AboutMedical LiteraturePublications

Hormones in Wellness and Disease Prevention

Keywords: Hormones, Estrogen, Progesterone, Testosterone, Thyroid, Growth hormone, Wellness, Prevention

The study and use of hormones have long been the domains of endocrinology, which is primarily focused on the pathologic phenomena encountered in the human body as they relate to hormones. No specific field in medicine has been designated to study and analyze the effects of hormones on wellness and disease prevention. As the field of wellness and disease prevention expands rapidly, it behooves the primary care practitioner, the first physician contact between the patient and the health care system, to become conversant and comfortable with hormone treatments as they relate to wellness and disease prevention.

Extensive scientific literature addresses the crucial role hormones play in the physiologic processes that maintain homeostasis. Much controversy surrounds the clinical use of various hormone therapies to support and maintain these processes in the aging patient. This article attempts to clarify some of the confusion and controversy surrounding estrogen, progesterone, testosterone, growth hormone, and thyroid hormones and discuss their roles as supported by the present state of evidence in disease prevention and aging as they apply to the primary care practice.

Hormones represent specific proteins produced by the human endocrine organs: pituitary, adrenals, thyroid, testes, and ovaries. Our focus is limited to estrogen, progesterone, testosterone, growth hormone, and thyroid. In health, all hormones are individually and wholly integral participants to the maintenance of cellular function and homeostasis. Hormone levels undergo diurnal variation and levels change in response to our environment, thought processes, stress levels, and food intake. Environmental toxins, medications, and pollutants also significantly affect hormone balance.

With the aging process, hormone levels decrease naturally. As these levels decline, problems with health maintenance arise. The diminution in hormone levels that occurs as a result of aging may or may not be compounded by concomitant disease states and environmental factors. In this article, we discuss age-related hormone loss and supplementation therapies for age-related hormonal deficiencies as possible firstline therapeutic modalities to be considered in our search to improve quality of life, prevent chronic illnesses, and maintain wellness.

Estrogen, Progesterone, Testosterone

Scientists have determined the existence of three true end-organ sex hormones: estrogen, progesterone, and testosterone. Both men and women have all three hormones, although levels and ratios of these hormones vary according to gender.

Estrogen and progesterone are the dominant hormones in women. We are often faced with the misconception that estrogen, progesterone, and testosterone act independently of one another. Without fully understanding the inseparable nature of the interaction between all sex hormones, we cannot solve the problems caused by imbalances in their individual levels and the symptoms these imbalances cause.

Estrogen is made in the ovaries, the corpus luteum, adrenal glands, and fat cells. Estrogen is not one big molecule; rather, it is a group of molecules. In humans, the three main identified estrogen molecules are estriol, estradiol, and estrone.

Estradiol is the most active form of estrogen made by the ovaries, adrenals, and fat cells postmenopause. Estradiol directly affects a wide range of cellular functions, as estrogen receptors are ubiquitous.

Estriol is the weakest of estrogens. Estriol is primarily manufactured during pregnancy by the placenta. It attaches to cell receptors affecting hair, nails, and skin. Recorded data on estriol’s function demonstrate that estriol’s effects are limited mainly to the vaginal walls with a little effect on the heart and bones in nonpregnant women. In the nonpregnant, young, and premenopausal woman, estriol is made in the liver in small doses. Studies on the use of estriol in menopausal women and women with multiple sclerosis have demonstrated promising results.

Estrone is manufactured in fat cells after menopause primarily from testosterone derivatives (androstenedione). Estrone levels tend to rise after menopause and the increase in estrone has been implicated in an increased incidence of breast tumors but most data have been obtained from animal studies. Overweight older women have high circulating levels of estrone.

When the scientific and lay communities refer to estrogen, they typically refer to its three components as one. At times, this oversimplification leads to errors in separating the individual function of the estrogens, particularly when discussing the differences between estrogen preparations used as hormone-replacement therapy available on the market. Although their actions are perceived and often recorded as one, the component molecules of estrogen have different potencies and effects.

During the aging process, the ovaries stop producing estrogen on a regular basis. Thereafter the main source of estrogen is from the adrenal glands, primarily in the form of estrone. The body transforms unused testosterone into estrogen (primarily estrone) and releases estrogen stored in fat cells.

Estrogen and progesterone are antagonists. Their actions are designed to balance each other and keep each other in check. We cannot live in a healthy state without hormonal balance. At no time do hormones act independently under normal circumstances in healthy bodies. For example, estrogen increases cell proliferation in the endometrium, while progesterone inhibits cell proliferation. Without progesterone, endometrial hyperplasia occurs in the uterus.

Progesterone is manufactured primarily by the corpus luteum (the follicle transformed after ovulation) and also to a small degree by the adrenals. In the ovary, progesterone production is activated at ovulation (15 days before the next menstruation), stimulated by the release of luteinizing hormone from the pituitary gland and is crucial to the survival of the ovum once fertilized. When pregnancy occurs, progesterone production increases rapidly and its manufacture is taken over by the placenta. If a woman does not get pregnant, the corpus luteum involutes and progesterone production diminishes and eventually disappears in parallel with estrogen production, heralding menstruation.

Progesterone is a precursor to most sex hormones, including estrogen in the ovaries, testosterone, all androgens, and other adrenal hormones, making it an extremely important hormone for reasons far beyond its role as a sex hormone. Progesterone in the breast and uterus counteracts the stimulation of cell growth, which is a direct action of estrogen. It accomplishes this action by activating the progesterone receptor, which in turn, down-regulates the estrogen receptor. Because progesterone suppresses estrogen-driven cell proliferation, progesterone in the natural state helps keep breast cell growth in healthy balance.

Estrogen and Progesterone: Nomenclature, Commercial Availability

Among the medications approved by the Food and Drug Administration (FDA) for hormone therapy are two classes of sex steroid hormones: estrogens and progestogens (which broadly include progesterone and progestogens or progestagens, also referred to as ‘‘progestins’’ or ‘‘progestational agents’’). For better clarification, these medications must further be divided into two groups: (1) bioidentical hormones with molecular structure identical to that of the human hormones and (2) preparations with molecular structures different from that of human hormones (nonidentical). The molecular difference between these two types of hormone formulations affect their actions in the human body.

In 2001, a literature review by Stanczyk scrutinized the various estrogen preparations available on the market. The investigator noted that the scarcity of comparative pharmacokinetic information between various formulas of estrogens created a void in our knowledge of their differential effects and thus hindered our ability to serve the patient. He encouraged comparative studies to help determine the best type of estrogen to be used as therapeutic options to enable individualized treatments and approaches that would fit each woman’s risk profile and personal preference.

Hormone Preparations with Molecular Formulas Unlike Those of Human Hormones

Hormone preparations that are molecularly different from human hormones are the most commonly used and marketed hormone-replacement therapy in the United States. They are commonly referred to in the popular literature as synthetic estrogens or pregnant horse urine estrogens. The most popular estrogenic preparations in this category include such oral estrogens as conjugated equine estrogen (Premarin), esterified estrogen (Estaratab, Menest, Cenestin), estrone sulfate (Ogen), and ethinyl estradiol (Estinyl); and such vaginal creams as estropipate (Ogen) and dienestrol (Ortho-dienestrol).

Progestins

Progestins, which include drug formulations that are also molecularly different from those for human progesterone, were developed to balance the endometrial hypertrophy associated with the use of unopposed conjugated estrogens on the uterus. Progestins are chemical compounds manufactured with two types of primary characteristics: androgenic and nonandrogenic properties. Progestins are manufactured in the laboratory and are not extracted from any known animal sources. They include medroxyprogesterone (Provera, Amen, Cycrin), norethindrone (Micronor, Norlutin), and norethindrone acetate (Norlutate).

Combination Products

Combination products contain combinations of both estrogenic and progestogenic compounds. Some include one hormone that is molecularly identical to human hormones and one that is not, while some contain both the estrogen and the progestogens that are molecularly different from human estrogen and progesterone. They include conjugated estrogen (nonidentical) and synthetic progestin (nonidentical) (Prempro, Premphase);17-beta-estradiol (bioidentical) and norgestimate (nonidentical) (Orthopresfest); ethinyl stradiol (nonidentical) and norethindrone acetate (nonidentical) (FemHRT); and esterified estrogens (nonidentical) and methyltestosterone (nonidentical) (Estratest).

Bioidentical Hormone Preparations

Bioidentical hormones are manufactured to be molecularly identical to hormones found in the human body. Bioidentical preparations include estradiol, estriol, progesterone, and testosterone. Bioidentical hormones are available both in commercial and compounded forms. Bioidentical hormones are not a marketing term. The term has been used for more than a decade in the inserts to all FDA-approved commercial hormone preparations that contain hormones molecularly identical to human hormones. Commercially and compounded available bioidentical hormone preparations include:

  • 17-Beta estradiol (Alora, Climara, Esclim, Estrace)
  • 17-Beta estradiol patches (FemPatch, Vivelle-Dot, Vivelle, Estraderm)
  • Estradiol transdermal spray (Evamist)
  • Progesterone in peanut oil capsule (Prometrium)
  • Progesterone vaginal gel (Crinone)
  • Micronized progesterone in various compounded forms (capsules, troches, transdermal creams, vaginal suppositories)
  • Combinations of estradiol and progesterone in compounded formulations as above
  • Combinations of estradiol, estriol, and progesterone in compounded formulations as above

Beyond the commercial bioidentical hormone formulations, individually compounded preparations of bioidentical hormones are prepared in compounding pharmacies or laboratories (some are FDA approved; all are regulated by the state they operate in) on an individualized basis as prescribed by a physician. These products contain the same active estrogens, progesterone, and testosterone as those found in the commercial preparations listed above. The difference is that they are individually mixed in tablet, capsule, troches, gels, or creams to the specifications of the prescribing physician for the individual patient. Unlike the commercial preparations, compounded hormone preparations are not manufactured on a large scale and can only be produced for individual patients as prescribed by a physician or other licensed practitioner, depending on the particular state rules.

In a recent review of bioidentical hormones in menopause, Boothby and colleagues reviewed only the compounded formulations of bioidentical. The investigators made no mention of the commercially available bioidentical hormones. This omission inadvertently perpetuated the confusion, credibility, and even existence of bioidentical hormones in FDA-approved commercially available preparations.

Much of the confusion surrounding estrogen and progesterone formulations comes from the lack of clear distinction between their molecular formulas, the lack of focus on their different effects in the human body, and the use of nonspecific nomenclature when referring to estrogen and progesterone regardless of formulaic or activity differences.

The molecular differences between bioidentical and nonhuman identical hormone preparations are illustrated in (Fig. 1).

Controversy

The differences in behavior of various hormone formulations in vivo and vitro are directly connected to the differences in molecular structure as described in the scientific literature. As early as 1976, scientific data demonstrating the safety of bioidentical hormones appeared in the conventional medical literature. Reports of increased risk of endometrial and breast carcinoma among users of synthetic conjugated estrogens also appeared in the scientific literature.

By January 1978, the Journal of the American Geriatrics Society addressed the growing concern that treatment with exogenous estrogen alone causes cancer and reported on progestogen as the solution. Adding small doses of a progestogen to either estradiol or conjugated estrogen in a cycled manner was determined to be a safe solution to the concern of increased carcinogenicity found with the use of unopposed estrogen. It is noteworthy that, in 1983, the options for treatment studied included bioidentical estradiol and conjugated estrogens with medroxyprogesterone. The stated goal of the treatment was to help women feel better as they aged and ‘‘not to harm’’ them in the process.

As early as 1980 and continuing into the recent literature, untoward side effects of synthetic progestins, such as thrombotic phenomena; breast tissue cell hyperplastic changes; and cardiovascular, cholesterol, carbohydrate, and lipid metabolism changes, prompted more research into bioidentical (micronized) progesterone as a safer option. An article in the British Medical Journal in March 1980 noted: ‘‘Clinically, oral bioidentical progesterone may be of value when synthetic progestogens have caused adverse symptoms that necessitate stopping treatment."

Recommendations for the use of bioidentical progesterone as a safer alternative were found in the medical literature from Europe as well as the United States throughout the early 1980s.

In the 1980s and early 1990s, research scientists expressed concern that the synthetic progestins in hormone therapy could increase the risk of breast cancer. About this same time, the scientific literature was replete with studies of safer alternatives in the form of bioidentical estradiol and progesterone, as well as studies comparing bioidenticals to the synthetic hormones and comparing various methods of administration with transdermal method of administration demonstrating the most promise in the area of safety and efficacy. Examples of such scientific literature included an article by Foidart and colleagues, who demonstrated that estradiol and progesterone had less proliferative effects on breast tissue cancer cell lines than did progestins and conjugated estrogens. Franke and Vermes showed that progesterone-induced apoptosis in breast cancer cell lines that were conversely stimulated by synthetic progestins and other androgenic progestins. Place and colleagues conducted a double-blind comparison of estradiol in transdermal form and Premarin that demonstrated improved relief of postmenopausal symptoms in the patient group on estradiol with no side effects. Riis and colleagues, in a double-blind clinical controlled study, demonstrated that bioidentical estradiol and micronized progesterone helped improve bone density in postmenopausal women. Moorjani and colleagues reported on the improved lipoprotein profile in patients receiving oral bioidentical estrogen with progesterone over those on progestins with androgenic action.

Screenshot 2020-10-20 162957.png

Fig. 1. The molecular formulas of various types of progestagens and estrogens. (Adapted from United States Pharmacopeia; and United States National Formulary.)

Notably, the Postmenopausal Estrogen/Progestin Interventions (PEPI) trial, a longterm randomized trial of hormone-replacement therapy, compared multiple effects, including cardiovascular effects, of both synthetic progestins and micronized progesterone in combination with conjugated equine estrogen. The PEPI trial confirmed that over the course of 3 years, oral conjugated estrogen taken alone or with synthetic progestins or micronized progesterone was associated with clinically significant improvement in lipoprotein profile and lowered fibrinogen levels. PEPI also demonstrated significant losses in high-density lipoprotein cholesterol when synthetic progestin was added (significantly reducing the beneficial effects of estrogen). However, when bioidentical progesterone was added, there appeared to be statistically significant endometrial sparing and the bulk of estrogen’s favorable effects on risk factors, including high-density lipoprotein cholesterol, were also preserved.

In 1994, the National Institutes of Health began the Women’s Health Initiative (WHI), a large-scale prospective double-blind placebo-controlled study. The goal of the study was to evaluate the long-term effect of hormone-replacement therapy versus placebo in the prevention of heart disease, osteoporosis, cancer, and strokes in postmenopausal women. The only form of hormone-replacement therapy used in the study was conjugated equine estrogens (conjugated estrogen [Premarin]) and medroxyprogesterone (synthetic progestins [Provera]). Unfortunately, the WHI did not include a bioidentical arm even though bioidentical hormone usage and statistically significant studies consistently demonstrated positive results and sustainable safety and efficacy records for this therapeutic modality.

Studies comparing the effectiveness and safety of different methods of administration (oral versus transdermal or vaginal), the use of synthetic versus bioidentical replacement, and the use of estrogen only versus combined estrogen and progesterone have raised more questions about the logic and safety of using conjugated estrogen and synthetic progestins in our patients. Large-scale studies have been conducted in Europe where bioidentical hormone replacement therapy is the main type of hormone supplementation in menopausal women. These studies repeatedly demonstrated effective elimination of menopausal symptoms and a lack of long-term negative side effects with the use of bioidentical preparations. Foidart and colleagues showed in a small study that, within 14 days, exposure to progesterone reduced the estradiol-induced proliferation of the breast epithelial cells in vivo in 40 postmenopausal women. E3N is a large prospective French cohort study that investigated breast cancer risk factors in 98,997 women born between 1925 and 1950. The data were analyzed every 2 years and the conclusion emerged that micronized progesterone regimens, compared to synthetic progestin regimens, were associated with significantly lower breast cancer risks. Additionally, women who took the hormone-replacement therapy consistently were at lower risk than women who took the hormones occasionally. De Lignie`res and colleagues reported the results of an 8.9-year study of a cohort of 3175 postmenopausal women using mainly transdermal estradiol and progesterone. No increased risk of breast cancer was found (risk ratio [RR] of breast cancer per year of use was 1.005). Stahlberg and colleagues reported on the Danish Nurse Cohort Study commenced in 1993, which followed women aged 45 and above. The highest risk of cancer was found in the women who used continuous combined estrogen with synthetic progestin. Nelson reviewed the studies that evaluated the short-term effectiveness of conjugated estrogen and estradiol as treatments for relief of hot flashes. The conclusion was that they both have comparable short-term effects. The overarching problem with conjugated estrogen is the long-term increased risk of breast cancer, stroke, and myocardial infarction, which was proven by the WHI initiative.

This situation leaves us with the very important knowledge that hormonereplacement therapy is an important tool in wellness and prevention. The type of hormone therapies we choose for our patients is what makes the difference and must be carefully considered.

Risks/Benefits

Scientific reviews of the pharmacology and action of progestins demonstrate that all progestins and progestogens are not created equal, and their action varies significantly according to their molecular structure. In the studies reviewed, bioidentical progesterone proved to be safer and more effective in all trials that involved its usage and numerous studies have shown that any estrogen (conjugated estrogen or bioidentical estradiol) combined with synthetic progestin doubles the risk of breast cancer. Unlike synthetic progestins, bioidentical progesterone has been shown to have a consistently beneficial effect on breast cell proliferation. The E3N and Danish Nurses studies, which address large populations taking various types of hormone-replacement therapy for more than 5 years, did not find progesterone to be an increased risk factor for breast cancer while progestin was. When estradiol was used in studies that evaluated its effectiveness in relieving menopausal symptoms, including hot flashes, night sweats, insomnia, and mood swings, and in improving sleep patterns and lipid profiles, the results were consistently positive.

The WHI study came to an abrupt halt in July 2002 primarily because the interim data demonstrated increased risk of myocardial infarction, stroke, and breast cancer in the conjugated estrogen and synthetic progestin arm of the study. Since that time, the suggestions to use hormone-replacement therapy in menopausal women has raised fears, doubts, and confusion. Millions of women, exposed to the media frenzy caused by the WHI’s unsettling results, abruptly stopped taking their hormone therapies at the advice of their physician and on their own. This situation required physicians to rethink hormone-replacement therapy and to look at other options for relief. Much time and effort has been spent on reevaluating the results of the WHI. This reexamination has brought to light many questions about the validity of the findings and soundness of the study. Despite questions raised about the validity of the WHI study, the study itself still provides grounds for caution. The use of synthetic estrogen and progestin replacement remains questionable at best.

Even though the only long-term study on hormone-replacement therapy in the United States was conducted on synthetic hormones and the data clearly established increased risk of cancers and strokes with the use of conjugated estrogen and progestins, hormone therapies are still the most effective therapeutic modalities for the elimination of symptoms of menopause and should be considered an integral part of the overall well-being of the aging woman. While in the short term, the type of hormones used may or may not be as significant as in the long run, the question is: What are the best options for the short and long terms for the women we treat?

An epidemiologic review of the rise in incidence in breast cancer in 1990 looked at the receptor status and the relationship to stage. Of interest is the fact that the investigator found that the incidence in older women increased and the cancers were more likely to be estrogen receptor positive. These cancers carry better prognosis because they tend to grow more slowly and are sensitive to hormonal manipulation. This information is useful for the primary care physician when deciding therapeutic course of action over the long term.

Subsequent to the discontinuation of the WHI study, hormones that are synthetic and molecularly dissimilar to human hormones can no longer be prescribed without hesitation. A growing number of physicians involved in prevention and wellness, in response to concerns raised by the WHI and to requests and demands from patients, have created study groups and forums within alternative and integrative medical organizations, have written books, and are conducting seminars sharing their clinical experience and research data on the use of bioidentical formulations of estrogen and progesterone. Risks associated with the use of conjugated estrogen and progestins, including the increased risks of breast cancer and cardiovascular events have not been reported with the use of bioidentical hormones.

Based on the extensive scientific data we have reviewed for this article, it is unclear whether any absolute circumstance calls for synthetic versions of hormonereplacement therapy and such use appears unwise. Given the easy commercial availability of bioidentical formulations and the lack of negative data on these hormones, primary care physicians can easily access them for their patients. When faced with the need to treat a woman with hot flashes, night sweats, insomnia, mood changes, loss of libido, and other symptoms of menopause, the primary care physician must choose wisely the safest and most effective way of improving the quality of life for the patient. While further long-term randomized trials would be helpful to quantify the difference in RRs between synthetic and bioidentical hormone replacement over the long term, the current state of evidence demonstrates bioidentical hormones as a safe and effective option to be considered separate and distinct from its synthetic counterparts.

Testosterone

Female

Although estrogen remains the central female hormone most frequently used in both wellness and disease prevention, much less controversy surrounds the use of testosterone in women, though the evidence either supporting or discouraging its use is scarce. Nicknamed ‘‘the hormone of desire’’ and promoted in the popular media as the rescuer from the plight of decreasing libido in aging women, testosterone has gained rapid acceptance in the prevention and wellness arenas at a time when controversy and confusion surround estrogen and progesterone therapies.

Testosterone is produced by the ovaries and adrenals in young women in low doses (free testosterone levels range between 2–8 pg/mL). The bulk of the present research on the use of testosterone has been conducted on women with surgical menopause, hypopituitarianism, anorexia nervosa, and primary adrenal insufficiency; patients with HIV and low body weight;67 and patients with glucocorticoid- and oral contraceptive– induced suppression of endogenous androgens. There has been little if any formal study on testosterone use in normal aging in women.

Benefits

Musclemass: The addition of testosterone to conjugated estrogen results in an increase in fat-free body mass and mitigates central fat deposition associated with estrogen use. In a double-blind placebo-controlled small study of androgen-deficient women, testosterone replacement demonstrably increased thigh muscle mass as measured by CT scanning. The data is very limited and its value and usefulness on large populations unknown. Further evaluation and research must be conducted as we address the possibility of usage of testosterone in the aging female to help improve muscle mass and decrease central adiposity.

Libido: Loss of libido in the aging female is the most common complaint that leads physicians to consider testosterone deficiency as a possible cause and the main consideration for treatment with testosterone. Multiple factors directly affect sexual inclination. Poor relationship status, self-image issues, multiple medications and their side effects, other stress factors, aging, and concurrent chronic or acute illnesses are some of the most frequently encountered deterrents of sex drive. Many of these factors cannot be altered, and all factors should be taken into account. Even so, testosterone appears to be effective in offsetting some of the effects from these factors, leading primary care physicians involved in integrative and wellness practices to make testosterone supplementation more popular.

Lack of training in the area of loss of libido and lack of concrete diagnostic criteria have created difficulties for the primary practitioner when attempting to address this problem. While circulating testosterone levels are not very helpful in diagnosing low testosterone as the cause for loss of libido, it may be helpful to keep in mind that premenopausal women have a range of 20 to 75 ng/dL total testosterone while postmenopausal women can present with values as low as 5 to 10 ng/dL. Because we rarely have comparative levels of testosterone on a patient before they come in with the complaint, it is almost impossible to determine whether the testosterone levels correlate in any way with the appearance of symptoms.

The seminal study on impaired sexual function improvement with supplemental testosterone comes from oophorectomized women. Seventy-five women 31 to 56 years old postoophorectomy and -hysterectomy were randomly assigned to receive conjugated estrogen and various doses of transdermal testosterone. The women who received the higher dose of testosterone reported a two- to threefold increase in sexual desire, masturbation, sexual intercourse, and sense of positive well-being as compared with placebo or conjugated estrogen alone.

Breast cancer Acting through androgen receptors, testosterone opposes estradiolinduced proliferation of human breast cell lines. Cases where endogenous testosterone levels are elevated, such as with polycystic ovary syndrome, are associated with breast tissue atrophy and a decreased risk of breast cancer. There are, however, conflicting data on the potential role of supplemental testosterone in the development of breast cancer and under no circumstances should testosterone be given without regular follow-ups.

Testosterone replacement considerations

Variation in dosing, method of administration, and duration of treatment are important determinants of safety and efficacy. To date, the medical literature contains little data on this topic. One is left with a smattering of information to help the patient rely on hopeful but dubious information obtained on the Internet and from popular literature.

Under these circumstances, a growing number of physicians involved with menopausal women’s wellness are using testosterone supplementation to provide improvement in libido and mood simply based on clinical findings and blood levels. A popular literature book The Hormone Of Desire by Susan Rako, MD, published in 1999, was followed by hundreds of articles in popular science that led to the rise of testosterone supplementation as a potentially helpful resource in the plight of aging women.

Formulations

Testosterone formulations include testosterone gel (Androgel), which is not FDAapproved for women, and various compounded formulations of testosterone in cream, subcutaneous pellets, oral, and sublingual forms. In summary, though treatment with testosterone in the aging woman is gaining popularity, there is a definitive need for studies specific to this population to evaluate the safety and efficacy of testosterone as a therapeutic modality for postmenopausal women, as well as for younger women with loss of libido, to define its best use in prevention and wellness. Studies are needed to help determine the safest and most efficacious methods for aging females to use testosterone.

Male

Testosterone is the primary androgen produced by the testes and it plays an essential role in the health of the male. Beyond determining the male sex characteristics, testosterone is a determinant of muscle strength, bone mass, libido, potency, and spermatogenesis.

Androgen deficiency

Androgen deficiency includes but is not limited to symptoms of decreased body hair, reduction in muscle mass and strength, increase in fat mass, decreased hematocrit, decreased libido, erectile dysfunction, infertility, osteoporosis, depression, and mood changes. Androgen deficiency may occur secondary to testicular or pelvic trauma or surgical removal, hypogonatropic hypogonadism, or with normal aging.

The normal aging process leads to adult hypogonadism with a decrease in levels of testosterone with age and the development of some or all of the symptoms enumerated above. The condition of androgen deficiency in aging is also known as andropause.

Androgen deficiency or hypogonadism is the result of subnormal production of testosterone by the testes. Its prevalence in healthy males over the age of 40 is demonstrated in observational studies, but there is no agreed upon blood level that defines deficiency.

Common causes of hypogonadism include but are not limited to:

Primary testicular failure

  • Klinefelter syndrome
  • Cryptorchidism
  • Orchitis
  • Trauma
  • HIV/AIDS
  • Myotonic muscular deficiency
  • Retroperitoneal fibrosis
  • Aging

Hypogonadotropic hypogonadism

  • Kallman syndrome
  • Prader-Willi syndrome
  • Idiopathic hypopituitarism
  • Pituitary tumors
  • Suprasellar tumors
  • Hemochromatosis
  • Inflammatory, traumatic, vascular lesions of pituitary and hypothalamus
  • Obesity
  • Severe chronic illnesses
  • Medication
  • Andropause

The risk of having low testosterone levels is significantly higher in men with hypertension (RR 1.84), hyperlipidemia (RR 1.47), diabetes (RR 2.09), obesity (RR 2.38) and asthma or chronic obstructive pulmonary disease (RR 1.40) than in men without these conditions. The prevalence of hypogonadism (defined as a total testosterone level below 300ng/dL) in 2162 men aged 45 years or older presenting to primary care offices was 38.7% in a study by Mulligan and colleagues.

Controversy

Perhaps the most significant controversy related to testosterone is the debate over its role in prostate health. For more than 60 years, traditional medical wisdom regarded testosterone as a significant risk factor for prostate hypertrophy and assumed that high testosterone levels served as fuel for prostate cancer. Hormone blockade and or estrogen therapy are still standard of care for prostate cancer therapy even today. Clinicians have hesitated to treat aging males with testosterone because of the belief that high levels of testosterone cause prostate cancer or speed up its growth. More than a decade ago, Shippen, Fryer, and Wright took the view that testosterone is actually protective and should be used. A ground-breaking study released in November 2007 provided a whole new set of data and a new perspective on testosterone. The results of this large-scale prospective study revealed that high endogenous levels of testosterone are associated with low mortality from all causes. The study suggests that low testosterone may be a predictive marker for those at high risk of cardiovascular disease.

Shores and colleagues79 investigated the correlation between testosterone levels (defined as total testosterone <250 ng/dL or free testosterone <0.75 ng/dL) and mortality in 858 males followed for up to 8 years. The results demonstrated that men with low circulating levels of testosterone had an 88% increased risk of mortality.

Benefits

Cardiovascular: Experimental studies suggest that androgens induce coronary vasodilatation. A placebo-controlled double-blind (PCDB) study performed in the United Kingdom followed 46 men with stable angina randomized to receive either a 5-mg testosterone patch or placebo in addition to their current medicines for 12 weeks. Both groups were then monitored for changes in treadmill exercise time before the onset of myocardial ischemia. The results of the treatment group compared with the placebo group were statistically significant (22% improvement in exercise time before onset of ST depression) without effect on prostate-specific antigen (PSA), hemoglobin, lipids, or coagulation profile during the duration of the study. Low-dose supplemental testosterone treatment in men with chronic stable angina increased exercise time preceding induced myocardial ischemia as defined by ST depression on EKG. Testosterone replacement therapy has also been proven to reduce insulin resistance, visceral adiposity, and cardiovascular risk. Additionally, a relatively low testosterone, independent of adiposity, is a risk factor for insulin resistance and type II diabetes and vice versa (insulin resistance and diabetes mellitus II are risk factors for low testosterone).

Anemia: Anemia is a frequent feature of male hypogonadism and antiandrogenic therapies. In a study that evaluated hemoglobin levels in 905 persons 65 years or older, of which 31 men and 57 women had anemia, hemoglobin levels were evaluated after 3 years. The participants were patients without cancer, renal insufficiency, or antiandrogenic treatments. Statistical evaluation of the results showed that older men and women with low testosterone levels had a higher risk of anemia.

Mood and quality of life: There is a compelling need for therapies that prevent Alzheimer’s disease, defer its onset, slow its progression, and alleviate its symptoms. In a study that evaluated the effects of testosterone therapy on cognition, neuropsychiatric symptoms, and quality of life in male patients with Alzheimer’s disease and healthy elderly men, 16 male patients with Alzheimer’s disease and 22 healthy male controls were treated with testosterone and a placebo gel daily. Patients receiving testosterone had significant improvement in quality-of-life scores and the treatment was well tolerated. Testosterone had minimal effects on cognition and the treated group showed more numerical improvement and less decline in visuospatial functions.

Osteoporosis and musculoskeletal: Untreated hypogonadism is a prominent cause of osteoporosis in men and bone mineral density significantly increases with testosterone treatment. Older men are as responsive to the anabolic effects of testosterone as young men. Testosterone induces skeletal muscle hypertrophy that leads to improved muscle strength in the leg as demonstrated in this study. A reciprocal change in lean and fat mass is observed but further studies are needed to determine the exact mechanism of change and the therapeutic doses needed for older men to obtain optimal results with minimum side effects.

Libido and sexual function: Treatment with testosterone improved sexual function in hypogonadal males in this very small study as measured by frequency and duration of erection and frequency of ejaculation. More studies in this important area must be undertaken to provide much-needed information. Perceived risks associated with testosterone treatments and its abuse in the areas of athletic enhancement have caused much confusion without scientific basis.

Risks

Prostate cancer: The connection between higher testosterone levels and growth of prostate cancer originated in 1941 with the publication of two papers by Huggins and colleagues. The data reported were based on one patient and, despite 67 years of subsequent studies that failed to establish scientific support for this theory, we are still faced with reluctance to treat men with testosterone supplementation for fear of giving them prostate cancer or fueling prostate cancer already present at a subclinical or microscopic level.

More than 430,000 men were part of longitudinal studies over the course of the past 67 years, and no well-designed study has ever shown a direct correlation between total testosterone levels and prostate cancer. A 2007 review out of Harvard concluded that:

Although there is yet to be a large, long term, controlled study on the effect of TRT [testosterone replacement therapy] on PCa [prostate cancer] risk, it should be abundantly clear that raising T [testosterone] in hypogonadal men has little, if any, impact on PCa risk or growth in the short to medium term. The withholding of TRT in men because of fear of PCa risk or progression is no longer tenable in an age of evidence-based medicine, because neither evidence nor theory supports this position.

This article reviewed the state of the evidence and, based on the prospective longitudinal studies, concluded that ‘‘men who develop prostate cancer do not have higher baseline testosterone levels and men with higher testosterone levels are at no greater risk for developing prostate cancer than men with lower testosterone levels.’’

The primary care physician needs to address each patient individually and decide on the use of testosterone based on more than just testosterone levels or fear of prostate cancer. Follow-up with serial blood tests and PSAs is still an important part of the clinical follow-up and should be used for the protection of the patient.

Aromatase: One of the most important factors affecting testosterone levels in aging men is the enzyme aromatase, which is found in fat tissue. Aromatase converts testosterone into estrogen, thus changing the ratio of estrogen to testosterone. Men who have excessive body and abdominal fat are likely to have increased estrogen levels caused by aromatase activity. This condition has been linked to decreased insulin sensitivity and metabolic syndrome.

Diagnosis

When a history and symptoms of hypogonadism are clear, the diagnosis is relatively easy. However, often the patient presents with nonspecific history and symptoms and an unremarkable clinical history, making the diagnosis more difficult. Clinically, the typical adult hypogonadism patient is above 50, fatigued, has difficulty building muscle in spite of consistent workout regimen, complains of unexplained weight gain, may be mildly depressed, and may experience erectile dysfunction and loss of libido. In this clinical setting without diagnosable disease, the diagnosis of a relative age-related adult-onset hypogonadism is gaining popularity and treatment with testosterone is becoming more common in the integrative medicine and urology fields.

Thus, it becomes important for the primary care physician, who is the first line of diagnosis and treatment, to feel comfortable with the use of testosterone as a viable and safe short- and medium-term option in the therapeutic armamentarium of healthy aging and wellness preservation. Understanding and considering hypogonadism in every adult aging male is an integral part of prevention and wellness.

Primary testicular failure is associated with elevated follicle-stimulating hormone and luteinizing hormone levels. A baseline PSA and a complete blood cell count should be obtained before starting testosterone supplementation. Estrogen, progesterone, and dihydrotestosterone levels may also be of value.

There is no agreed total or free testosterone cut-off level to define testosterone deficiency. Total testosterone is the most common measure of androgen activity, but is a poor indicator of tissue activity, demonstrating little correlation with clinical status, and is an unreliable indicator of response to therapy.

Free testosterone is a more accurate indicator of hypogonadism, but normal ranges for total and free testosterone vary widely among laboratories, even among those using the same assay, and the reference ranges show little or no correlation to clinical findings. When testing the testosterone levels of a patient who is considering testosterone supplementation to maintain and improve wellness, it is unusual to have available prior testosterone levels when that patient was younger, healthier, and symptom free. Thus, a result that appears to be within normal range may not necessarily reflect what is normal for that particular patient. This situation must be taken into account since it emphasizes the importance of clinical assessment and patient involvement in the decision to treat.

The use of population-based statistically determined normal testing ranges is also limited by the fact that the average testosterone level in men today is less than the average level in men of the same age 15 years ago. This concerning fact is possibly due to environmental suppression of the hypothalamic-pituitary-testicular axisand may also be a contributing factor to diminished sperm counts and increased incidence of infertility.

Testosterone levels decrease with age and illness. Typically, men with hypogonadotropic hypogonadism have low plasma testosterone and luteinizing hormone levels. Prolactin levels should be checked if the total testosterone level is below 250 ng/dL to rule-out a pituitary tumor.

Fifty percent of circulating testosterone is bound to sex hormone–binding globulin, which directly affects free testosterone levels. Free testosterone levels can be obtained to clarify testosterone status. However, variations are greater among free testosterone assays than among total testosterone assays. Also, reference ranges are not as standardized for free testosterone assays as they are for total testosterone assays. When borderline levels of testosterone are found, or the clinical picture and the blood tests disagree, a low or low-normal free or total testosterone level may be used to support a clinical diagnosis of androgen deficiency, but should not be used to exclude it.

Treatment

Testosterone supplementation has gained popularity over the past 20 years. The benefits of testosterone supplementation include improved energy, greater muscle mass, increased stamina, greater strength, increased confidence, greater motivation, and enhanced libido.

Present formulations of testosterone include the following:

  • Testosterone gel (Androgel)
  • Testosterone patches (Androderm)
  • Compounded testosterone creams or gels
  • Injectable testosterone
  • Subcutaneous testosterone implants

Monitoring

While it is useful to follow PSA levels during the course of testosterone replacement and supplementation, it is more important to track the velocity PSA increase. There is often a slight bump, a rise above 4.0 ng/mL, or a sudden increase in PSA with the initiation of testosterone therapy, followed by a stable constant level. An increase in PSA more than 0.35 ng/mL per year warrants further evaluation and a referral to the urologist. While using testosterone in disease prevention and wellness is relatively new to the primary care field, it holds much promise and meets with much support and enthusiasm from patients. The data we reviewed and our clinical experience support the use of testosterone as a first-line hormone supplementation in the aging male. More research is needed to substantiate and define the parameters necessary for its long-term use.

For now, as the esteemed Dr. Morgantaler said:

... the diagnosis of androgen deficiency requires only an ear attuned to the characteristic symptoms and blood test providing evidence of reduced levels of total or free testosterone. Treatment provides an opportunity for gratifying results, for patients and clinicians alike.

Growth Hormone

As the proportion of aging people continues to rapidly rise, reducing the burden of age-related diseases becomes increasingly important in primary care. A controversial hormone that is center stage in the debate over the use of hormone therapies in prevention and wellness is growth hormone.

Growth hormone, a single-chain polypeptide produced in the pituitary gland, has a wide range of metabolic and cellular effects. Growth hormone plays an important role in the regulation of body composition, lipid profiles, tissue repair, cardiac and neuronal functioning, and maintenance of bone mineral density. Growth hormone is secreted in pulsatile fashion, especially during stage III and IV deep sleep. It acts on liver and other tissues to stimulate the production of insulinlike growth factors (IGFs), including IGF-1, which is also known as somatomedin C, and the production of IGF-binding proteins (IGFBPs), which also have direct cellular actions. The most abundant IGFBP is IGFBP-3.

A large percentage of growth hormone effects are mediated through IGF-1. Because of the pulsatile nature of growth hormone production and short half-life (20–50 minutes), routine serum growth hormone levels cannot be used to determine overall production. While there are many influences on the production of IGF-1, levels correlate with overall growth hormone production, are relatively stable in the serum, and are currently the best estimate of growth hormone production and effect. While a low IGF-1 is a strong indicator of abnormally low growth-hormone production, an IGF-1 level in the normal reference range does not rule out deficiency.

While there is considerable variation in growth hormone production among individuals of the same age, there is a progressive decline in average growth-hormone production and IFG-1 levels after age 20, with average levels declining by 30% to 60% by age 40 to 60, and by 50% to 80% after age 60. Low growth-hormone levels and production are associated with low quality of life as measured by numerous criteria, including the Nottingham Health Profile and the Psychologic General Well-Being Index.113,115–118 Gibney and colleagues119 reviewed 10 years of use of growth hormone in adult growth-hormone deficient patients and found it to be of significant benefit.

A large number of peer-reviewed research, including long-term randomized controlled trial data, has demonstrated that growth hormone replacement improves energy, strength, cardiac function, blood pressure, cholesterol levels, insulin sensitivity, cognitive function, immunity, psychologic well-being, decreases body fat, increases lean muscle, prevents and reverses heart disease, prevents and improves osteoporosis, and improves quality of life.

Controversy

Controversial issues regarding growth hormone supplementation include the use of growth hormone as a therapeutic modality for age-related deficiency; the accuracy and necessity of commonly used stimulation testing when considering growth hormone usage in well patients; the need for guidelines for safe and effective treatment; and potential side effects of treatment.

Diagnostic Testing

The diagnosis of growth hormone deficiency is difficult for a number of reasons. As discussed, random serum growth-hormone levels are not indicative of the overall growth hormone production and, while IGF-1 levels do correlate with overall growth hormone production, IGF-1 levels lack sensitivity to detect significant deficiency (IGF-1 levels are often in the normal range even if a significant deficiency exists).

With growth hormone stimulation testing, serum growth-hormone levels are measured after a variety of agents and protocols are used to stimulate the release of growth hormone from the pituitary. Such tests are often promoted as the means of differentiating growth hormone deficiency from normal state. Many endocrinologists believe the diagnosis of adult growth-hormone deficiency can only be made with the use of growth hormone stimulation testing. Such testing has proven to be inaccurate, highly variable, nonphysiologic, and lacking adequate sensitivity to detect relative growth-hormone deficiencies. The use of arbitrary cutoffs to define abnormality does not correlate with response to therapy. Studies demonstrate that using the same agent to perform stimulation tests multiple times on one patient do not consistently produce congruous results, thus bringing the usefulness of the test into question. Side effects of stimulation testing include significant hypotension, venous thrombosis, nausea, and vomiting. Deaths and neurologic damage have also been reported.

Because stimulation tests are clinically and physiologically unreliable, they are also unreliable for determining growth hormone deficiency. Currently the most appropriate means of diagnosing age-related growth-hormone deficiency is clinical recognition and a low-normal (below the mean) IGF-1 level.

Clinical Diagnosis

The adult age-related clinical syndrome of growth hormone deficiency includes increased fat mass, decreased muscle mass and strength, decreased bone density, elevated lipids, insulin resistance, decreased psychosocial well-being and depression, fatigue, increased social isolation, inability to handle stress, cardiovascular disease, memory decline, overall deterioration in quality of life, frailty, thin dry skin, increased wrinkles, and diminished exercise tolerance.

Clinicians commonly encounter these clinical symptoms in the aging patient. If considered appropriate by physician and patient, a 6-month therapeutic trial with growth hormone could be considered, dosed to keep IGF-1 levels in the upper quartile. Patients should be evaluated for symptomatic and metabolic improvements at a minimum at 3 and 6 months to decide if treatment should be continued.

Treatment

The treatment of age-related adult growth-hormone deficiency remains controversial even though the literature reports significant benefits from growth hormone supplementation. The main sources of concern associated with growth hormone replacement in somatopause include, in no particular order, significant cost of therapy from $250 to $1500 per month (depending on dose and manufacturer), side effects of water retention resulting in joint pain and carpal tunnel syndrome, temporary reduction in insulin sensitivity, and theoretic risk of cancer. Most short-term side effects are diminished with reduction in dose.

While there is a long-held theoretic belief of an increased risk of cancer, based on the growth hormone’s antiapoptotic and mitogenic effects, neither long-term nor short-term data support this theory. Conflicting data on the relationship between IGF-1 levels and the risk of cancer abound. Some frequently cited epidemiologic studies have found an increased correlation between elevated IGF-1 and breast, prostate, and colorectal cancers, while the majority of studies failed to document increased risk of cancer (or have shown a decreased risk) with increasing IGF-1 levels. In addition, one frequently cited study that did connect increased IGF-1 levels and cancer, by Chan and colleagues, is very controversial because the blood was stored for 5 to 15 years before it was tested. Also, IGF-1 levels in the highest quartile group were over three times the upper limit of normal for this age group, suggesting that IGF-1 in the patients studied may not have been measured accurately. Hankinson and colleagues found a trend for decreased risk of breast cancer in postmenopausal women with increased IGF-1 levels but an increased risk in premenopausal women. Palmqvist and colleagues reported increased association between IGF-1 and colon cancer, but a decreased risk of rectal cancer.

The secretion and regulation of IGF-1 is extremely complex and their reported association with cancer must also take into consideration numerous other potential confounding etiologic factors, whether environmental, nutritional, or other yet unidentified. Growth hormone stimulates the production of IGFBP-3, which has cancerprotective characteristics and may counteract increased risk of cancer associated with an increase in IGF-1, if present. There is evidence that tumors secrete IGF-1, which makes it a potential marker for cancer in some individuals and not necessarily a cause. Typical growth hormone supplementation for an age-related deficiency results in small increases in IGF-1 that remain in the normal age-matched references range, so risk would not be expected to be different than that for controls.

None of the long- and short-term studies have shown an increased risk of cancer, recurrent or de novo, with the use of growth hormone, and some of the studies have shown a decreased risk. Among these studies are studies on more than 19,000 children representing of 47,000 patient years of growth hormone treatment; a prospective study of 100 adult growth hormone–deficient patients followed for 1 to 4 years, a study of 910 children treated with growth hormone for 11 years, a study of 32 adults and children followed for up to 40 years treated with growth hormone (average 10.8 years); a study of 180 growth hormone–treated children followed for over 6 years with reduced cancer recurrence risk (RR 0.6); a prospective analysis of 289 growth hormone–deficient adults who, after 5 years of growth hormone therapy, showed lower risk of malignancy (RR 0.25) and decreased risk of myocardial infarction (RR 0.19) and early mortality (RR 0.22) compared with the untreated group.

In 2001, the consensus statement by the Growth Hormone Research Society noted that the data demonstrate that the concern for increasing the risk of cancer with the use of growth hormone is unfounded:

The current labeling for GH [growth hormone] states that active malignancy is a contraindication of GH treatment. There are, however, no data to support this labeling. Current knowledge does not warrant additional warning about cancer risk on the product label.

Supraphysiologic doses of growth hormone are shown to antagonize the effects of insulin. While short-term studies using large doses of growth hormone may potentially worsen insulin resistance, low physiologic doses of growth hormone have demonstrated improvement in insulin resistance and decreased risk of diabetes. If treatment is contemplated, low physiologic doses should be used to keep IGF-1 in the upper limit of normal.

In conclusion, aging adults have a relative deficiency of growth hormone and supplementation with growth hormone may be of significant benefit. A clinical diagnosis of growth hormone deficiency can be made with support of low-normal IGF-1 levels alone. Although no long-term studies have assessed side effects with low physiologic doses of growth hormone supplementation in somatopause, the studies we reviewed above have confirmed that low doses, titrated to keep IGF-1 levels in the upper limit of normal, are safe, well tolerated, and associated with a plethora of clinical benefits.

Treatment with growth hormone is presently limited to an affluent and highly motivated population. Cost and risk/benefit ratio over time must be taken into consideration. As our patients age, the challenge of maintaining quality of life for them becomes more difficult and must be considered in the design of future studies. For supplementation with growth hormone to become a first-line therapeutic option in the aging population, additional and more extensive randomized trials that evaluate results of growth hormone treatment in age-related deficiency must be undertaken, and cost factors must be addressed.

Thyroid

Hypothyroidism is a common disorder with inadequate amounts of thyroid hormone present at the cellular level. Typical symptoms include fatigue, weakness, weight gain, cold intolerance, muscle aches, headaches, decreased libido, depression, hair loss, and dry skin. Signs include edema, dry skin, pallor, hair loss, loss of temporal eyebrow hair, and cold extremities. Conditions associated with hypothyroidism include hypertension, atherosclerosis, hypercholesterolemia, hyperhomocysteinemia, menstrual irregularities, infertility, premenstrual syndrome, chronic fatigue syndrome, fibromyalgia, fibrocystic breasts, polycystic ovary syndrome, depression, diabetes, and insulin resistance.

There is a two- to threefold increase in the incidence of thyroid dysfunction with age, including overt and subclinical hypothyroidism (elevated thyrotropin with normal thyroxine and triiodothyronine levels). There is also an age-related decrease in thyroid function that results in diminished tissue thyroid levels and may result in clinically symptomatic hypothyroidism that is not detected with the standard use of thyrotropin, thyroxine, or triiodothyronine levels.

Historically, an elevated thyrotropin with normal thyroxine and triiodothyronine levels has been considered compensated or subclinical hypothyroidism and diagnosed as euthyroid with no requirement for treatment. A plethora of studies have, however, demonstrated that, in spite of the normal triiodothyronine and thyroxine values, subclinical and nondiagnosed hypothyroidism is often associated with significant symptoms and an increased risk of morbidity and mortality. In light of this, it has been proposed that the term subclinical hypothyroidism be replaced by the term mild thyroid failure (MTF).

The diagnosis of MTF is particularly important in the aging population in the areas of prevention and wellness. MTF is a treatable condition associated with increased cardiovascular risk and numerous signs and symptoms that might otherwise be attributed to ‘‘usual’’ signs and symptoms of aging, including fatigue, depression, memory loss, cognitive dysfunction, dry skin, constipation, leg cramps, cold intolerance, weakness, water retention, diminished sweating, weight gain, and diminished exercise tolerance. Significant improvements may occur with treatment.

Numerous studies have demonstrated increased cholesterol levels in patients with MTF. Thyroid replacement results in a significant reduction in the cholesterol levels.205–207 In addition to the increase in total and low-density cholesterol seen with MTF, endothelial dysfunction with impaired vasodilatation have also been demonstrated, further increasing the risk of cardiovascular events.

The Rotterdam study investigated the association between MTF and aortic atherosclerosis and myocardial infarction in 1149 menopausal women. After adjustment for multiple known coronary artery disease risk factors, the investigators found that MTF significantly increased the risk for arthrosclerosis (odds ratio 1.9) and myocardial infarction (odds ratio 3.1). This important study found that subclinical hypothyroidism was a greater risk for myocardial infarction than hypercholesterolemia, hypertension, smoking, or even diabetes, and that MTF was a contributing factor in 60% of the myocardial infarctions in the patients studied.

In a 20-year longitudinal study, Walsh and colleagues also examined the association between MTF, cardiovascular disease, and mortality in over 2000 individuals (approximately half men and half women) with a mean age of 50 years (age range 17–89). In this study, MTF was associated with a 2.2-fold increased risk of coronary artery disease and 1.5-fold increased risk of cardiovascular mortality after adjustment for multiple known cardiovascular risk factors.

Diagnostic Testing

Thyrotropin is considered the most sensitive marker of peripheral tissue levels of thyroid hormone, and it is widely assumed that thyrotropin levels within the normal range indicate the person is euthyroid. With significant physiologic stress, illness, inflammation and aging, however, there is demonstrable suppression of thyrotropin, making the thyrotropin test unreliable. With significant physiologic stress, illness, inflammation, and aging, tissue-specific alterations also reduce tissue triiodothyronine levels by reducing uptake of thyroxine into tissues and decreasing thyroxine-to-triiodothyronine conversion. The decreased serum thyroxine levels caused by the suppressed thyrotropin production is offset to varying degrees by the diminished uptake of thyroxine into the cell and the decreased thyroxine-to-triiodothyronine conversion. This situation tends to be misread as an indication of adequate tissue thyroid levels and makes thyroxine levels of little use, except in extreme cases.

With physiologic stress, inflammation, illness, and aging, the correlation between serum thyrotropin and thyroxine levels and peripheral thyroid activity no longer follows. Thyrotropin and thyroxine levels cannot be relied upon to detect diminished cellular triiodothyronine levels for aging patients and patients under stress. Instead of thyroxine normally converting intracellular to the active triiodothyronine in peripheral tissue, thyroxine is preferentially converted to reverse triiodothyronine. Serum reverse triiodothyronine levels may be useful because diminished cellular uptake of thyroxine, diminished thyroxine-to-triiodothyronine conversion, and diminished cellular triiodothyronine levels inversely correlate with serum reverse triiodothyronine levels.

When the physiologic stress or illness is acute and severe, the significantly diminished thyroid levels in the peripheral tissues no longer correlate with thyrotropin levels. This is termed nonthyroidal illness or euthyroid sick syndrome. In these cases, the thyrotropin level cannot be relied upon as an accurate measure of tissue thyroid effect. The same physiologic changes also occur with chronic physiologic stress, chronic illness, inflammation, calorie reduction, and aging. Changes can be metabolically significant and can cause serious symptoms. Treatment may be warranted despite normal thyrotropin and thyroxine levels. The use of thyroxine preparations in the treatment of nonthyroidal illness found in acute conditions, such as trauma, surgery, and sepsis, has shown little benefit. The ineffectiveness of thyroxine preparations in these cases is most likely due to the diminished use and uptake of thyroxine in these conditions. In contrast, treatment with triiodothyronine has proven quite beneficial in studies of severely ill patients, as well as in chronic conditions, which correlate well to the aging patient.

Similar to significant physiologic stress and illness, aging is associated with significant alterations in the hypothalamic-pituitary-thyroid axis that result in a reduction of thyrotropin levels (in contrast to MTF’s increase in thyrotropin) while tissue-specific alterations reduce the supply of triiodothyronine (via reduced thyroxine-to-triiodothyronine conversion and reduced uptake of thyroxine) to the body tissues.

With aging, as with nonthyroidal illness, thyrotropin and thyroxine are not indicative of tissue levels of triiodothyronine, making the interpretation of thyroid function tests increasingly complicated and difficult. Aging may be considered a chronic nonthyroidal illness leading to decrease in basal metabolic rate and reduction in thyrotropin and triiodothyronine levels without a significant decrease in thyroxine and free thyroxine (Fig. 2). Elevation in reverse triiodothyronine level is also seen as a consequence of diminished use of thyroxine, diminished thyroxine-to-triiodothyronine conversion, and diminished tissue levels of triiodothyronine. Another finding in the aging patient is the significantly reduced thyrotropin response to thyrotropin-releasing hormone that is similar to that found in severely ill patients with documented nonthyroidal illness.

Screenshot 2020-10-21 105917.png

Fig. 2. Age-dependent variations in (A) free thyroxine (FT4), (B) free triiodothyronine (FT3), and (C) thyrotropin (TSH). All healthy subjects in the study (groups A–C) were pooled for this analysis. (From Mariotti S, Barbesino G, Caturegli P, et al. Complex alteration of thyroid function in healthy centenarians. J Clin Endocrinol Met 1993;77(5):1132; with permission. Copyright ª 1993, The Endocrine Society.)

Further contributing to potential inaccuracies of standard thyroid testing in this population is the increasing incidence of systemic illness and the increased use of medications that directly affect thyroid function. In aging patients who present with symptoms consistent with hypothyroidism but have a normal thyrotropin and thyroxine level, obtaining free triiodothyronine, reverse triiodothyronine, and triiodothyronine/reverse-triiodothyronine ratios may help obtain a more accurate evaluation of tissue thyroid status and may be useful to predict those who may respond favorably to triiodothyronine supplementation.

The inaccuracy of thyrotropin and thyroxine levels in this potentially large group of individuals, including those with chronic physiologic stress, illness, and advancing age, has potentially profound implications. Studies that do not address the complex interactions of the aging thyroid and illness and use thyrotropin and thyroxine levels alone to determine thyroid status may be significantly flawed. With increasing knowledge of the complexities of thyroid function at the cellular level, it is becoming increasingly clear that the thyrotropin may not be as reliable a marker of tissue thyroid levels as once thought, especially with chronic physiologic stress, illness, inflammation, and aging. It is possible that many symptomatic patients with low tissue levels of active thyroid but normal thyrotropin and thyroxine levels would benefit from thyroid replacement both short and long term. Increasing evidence shows that thyroxine is not an optimal treatment for conditions associated with diminished use of thyroxine. Conversion of thyroxine to triiodothyronine (increased formation of reverse triiodothyronine) should lead the clinician to consider treatment with triiodothyronine.

Thyroid Preparations

Thyroid preparations include triiodothyronine (Cytomel); thyroxine (Synthroid, Levothyroxine); combinations of triiodothyronine, thyroxine; and compounded thyroid formulations (including thyroxine/triiodothyronine and timed-released triiodothyronine preparations).

Further studies are needed regarding the use of triiodothyronine preparations in the aging population and long-term outcomes based on treatment strategies that use improved methods for determining tissue thyroid levels instead of sole reliance on thyrotropin testing. With so much potential for inaccuracy in our present standard thyroid testing, the importance of additional or alternative methods for clinical assessment cannot be overemphasized. New methods of determining tissue levels of thyroid in the aging patient must be developed and used to better assess both short-term and long-term treatment effects and to help the primary practitioner assess tissue thyroid activity in the aging patient with symptoms and normal thyrotropin, thyroxine, and triiodothyronine levels.

Summary

In summary, we believe the well-informed use of hormones in wellness and disease prevention will result in symptomatic improvement and should be considered an integral part in the armamentarium of options we offer our patients. Definitions and testing of hormone deficiency that apply to illnesses do not apply to wellness and prevention and need to be reevaluated while we develop new treatment paradigms to best care for our patients. With the limited amount of research focused primarily on the areas of wellness and prevention, we must acknowledge the infinite number of variables that confound the results of every study. Ultimately we must focus on the individual patient and his or her need and that is the area where the doctor–patient relationship is of utmost importance and is the key to true prevention and wellness.

Resources

  1. Sicotte MD, Liva SM, Klutch R, et al. Treatment of multiple sclerosis with the pregnancy hormone estriol. Ann Neurol 2002;52(4):421–8.
  2. Mueck A, Seeger H, Wallwiener D. Comparison of the proliferative effects of estradiol and conjugated equine estrogens on human breast cancer cells and impact of continuous combined progestogen addition. Climacteric 2003;6: 221–7.
  3. Tourgeman D, Gentzchein E, Stanczyk F, et al. Serum and tissue hormone levels of vaginally and orally administered estradiol. Am J Obstet Gynecol 1999; 180(6 Part 1):1480–3.
  4. Lippert T, Seeger H, Mueck A. Pharmacology and toxicology of different estrogens. G Endodonzia 2001;15:26–33.
  5. Stanzcyk F. Estrogen used for replacement therapy in postmenopausal women. G Endodonzia 2001;15(4):17–25.
  6. Ribot C, Tremollieres F. Hormone replacement therapy in postmenopausal women. All the treatments are not the same. Gynecol Obstet Fertil 2007;35: 1–10. 7.
  7. Schindler A, Campagnoli C, Druckman R, et al. Classification and pharmacology of progestins. Maturitas 2003;46:S7–16.
  8. Smith D, Prentice R, Thompson D, et al. Association of exogenous estrogen and endometrial carcinoma. N Engl J Med 1975;293(23):1164–6.
  9. Ziel H, Finkle W. Increased risk of endometrial carcinoma among users of conjugated estrogens. N Engl J Med 1975;293(23):1167–70.
  10. Stanczyk FZ. All progestins are not created equal. Steroids 2003;68:879–90.
  11. Mack TM, Pike MC, Henderson BE, et al. Estrogens and endometrial cancer in a retirement community. MEJM 1976;294(23):1262–7.
  12. Foidart J, Colin C, Denoo X, et al. Estradiol and progesterone regulate the proliferation of human breast epithelial cells. Fertil Steril 1998;69(5):963–9.
  13. Boothby LA, Doering PL, Kipersztok S. Bioidentical hormone therapy: a review. Menopause 2004;11:356–67.
  14. Franke H, Vermes I. Differential effects of progestogens on breast cancer cell lines. Maturitas 2003;46:55–8.
  15. Druckman R. Progestins and their effects on the breast. Maturitas 2003;46: 59–69.
  16. Colditz G. Estrogen, estrogen plus progestin therapy, and risk of breast cancer. Clin Cancer Res 2005;11:909–17.
  17. Callantine M, Martin P, Bolding OT, et al. Micronined 17b-estradiol for oral estrogen therapy in menopausal women. The Am Col of Obstet Gynecol 1975;46: 37–41.
  18. Weiss N, Szekely D, Austin D. Increasing incidence of endometrial cancer in the United States. NEJM 1976;294(23):1259–62.
  19. Greenblatt R, Stoddard L. The estrogen-cancer controversy. J Am Geriatr Soc 1978;26(1):1–8.
  20. Whitehead M, Townsend P, Gill D, et al. Absorption and metabolism of oral progesterone. BMJ 1980;280(6217):811–27.
  21. Morville R, Dray F, Reynier J, et al. Biodisponibilite de la Progesterone Naturelle Administree Par Voie Orale. Journal De Gynecologie Obstetrique 1982;11(3): 355–63 [in French].
  22. Lane G, Siddle N, Ryder T, et al. Dose dependent effects of oral progesterone on the oestrogenised postmenopausal endometrium. BMJ 1983;287:1241–5.
  23. Chang K, Fournier S, Lee T, et al. Influences of percutaneous administration of estradiol and progesterone on human breast epithelial cell cycle in vivo. Fertil Steril 1995;63(4):785–91.
  24. Bergkvist L, Adami H, Persson I, et al. The risk of breast cancer after estrogen and estrogen-progestin replacement. NEJM 1989;321(5):293–7.
  25. Glass A, Hoover R. Rising incidence of breast cancer: relationship to stage and receptor status. Natl Can Inst 1990;82:693–6.
  26. Place V, Powers M, Schenkel L, et al. A double-blind comparative study of estraderm and premarin in the amelioration of postmenopausal symptoms. Am J Obstet Gynecol 1985;152(8):1092–9.
  27. Riis B, Thomsen K, Strom V, et al. The effect of percutaneous estradiol and natural progesterone on postmenopausal bone loss. Am J Obstet Gynecol 1987; 156(1):61–5.
  28. Maxson W, Hargrove J. Bioavailability of oral micronized progesterone. Fertil Steril 1985;44(5):622–6.
  29. Hargrove J, Maxson W, Wentz A, et al. Menopausal hormone replacement therapy with continuous daily oral micronized estradiol and progesterone. Obstet Gynecol 1989;73(4):606–12.
  30. Whitehead M, Fraser D, Schenkel L, et al. Transdermal administration of oestrogen/progesterone hormone replacement therapy. The Lancet 1990;335: 310–2.
  31. Moorjani S, Dupont A, Labrie F, et al. Changes in plasma lipoprotein and apolipoprotein composition in relation to oral versus percutaneous administration of estrogen alone or in cyclic association with utrogestan in menopausal women. J Clin Endocrinol Metab 1991;73(2):373–9.
  32. Erpecum K, van Berge Henegouwen G, Verschoor L, et al. Different hepatobiliary effects of oral and transdermal estradiol in postmenopausal women. Gastroenterology 1991;100:482–8.
  33. Nachtigall L. Emerging delivery systems for estrogen replacement: aspects of transdermal and oral delivery. Am J Obstet Gynecol 1995;173(3 Part 2):993–7.
  34. Writing group for the PEPI trial. Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The postmenopausal estrogen/progestin interventions (PEPI) trial. JAMA 1995;273:199–208.
  35. Thornton K, DeFronzo R, Sherwin R, et al. Micronized estradiol and progesterone: effects on carbohydrate metabolism in reproductive-age women. Society for Gynecologic Investigation 1995;2(4):643–52.
  36. Speroff L, Whitcomb R, Kempfert N, et al. Efficacy and local tolerance of a lowdose, 7-day matrix estradiol transdermal system in the treatment of menopausal vasomotor symptoms. Obstet Gynecol 1996;88(4):587–92.
  37. Evans S, Davie M. Low and conventional dose transdermal oestradiol are equally effective at preventing bone loss in spine and femur at all postmenopausal ages. Clin Endocrinol 1996;44:79–84.
  38. Good W, John V, Ramirez M, et al. Double-masked, multicenter study of an estradiol matrix transdermal delivery system (Alora) versus placebo in postmenopausal women experiencing menopausal symptoms. Clin Ther 1996;18:1093–105.
  39. Ross D, Cooper A, Davies J, et al. Randomized, double-blind, dose-ranging study of the endometrial effects of a vaginal progesterone gel in estrogentreated postmenopausal women. Am J Obstet Gynecol 1997;177(4):937–41.
  40. Stahlberg C, Pedersen A, Lynge E, et al. Increased risk of breast cancer following different regimens of hormone replacement therapy frequently used in Europe. Int J Cancer 2004;109:721–7.
  41. Nelson HD. Commonly used types of postmenopausal estrogen for treatment of hot flashes. JAMA 2004;291(13):1610–20.
  42. Fournier A, Berrino F, Riboli E, et al. Breast cancer risk in relation to different types of hormone replacement therapy in the E3N-EPIC cohort. Int J Cancer 2005;114:448–54.
  43. Fournier A, Berrino F, Clavel-Chapelon F. Unequal risks for breast cancer associated with different hormone replacement therapies: results from the E3N cohort study. Breast Cancer Res Treat 2008;107(1):103–11.
  44. De Lignie`res B, de Vathaire F, Fournier S, et al. Combined hormone replacement therapy and risk of breast cancer in a French cohort study of 3175 women. Climacteric 2002;5:332–40.
  45. Santen RJ. Risk of breast cancer with progestins: critical assessment of current data. Steroids 2003;68:953–64.
  46. Schairer C, Lubin J, Troisi R, et al. Menopausal estrogen and estrogen-progestin replacement therapy and breast cancer risk. JAMA 2000;283:485–91.
  47. Campagnali C, Abba C, Ambroggio S, et al. Breast cancer and hormone replacement therapy: putting the risk into perspective. Gynecol Endocrinol 2001;15:53–60.
  48. Lyytinen H, Pukkala E, Ylikorkala O. Breast cancer risk in postmenopausal women using estrogen-only therapy. Obstet Gynecol 2006;108(6):1354–60.
  49. Li C, Malone K, Porter P, et al. Relationship between long durations and different regimens of hormone therapy and risk of breast cancer. JAMA 2003;289(24): 3254–63.
  50. Schindler A. European Progestin Club. Differential effects of progestins. Maturitas 2003;46:S3–5.
  51. Grady D, Vittinghoff E, Lin F, et al. Effect of ultra-low-dose transdermal estradiol on breast density in postmenopausal women. Menopause J North Am Men Soc 2007;14(3):1–6.
  52. Simon JA, Bouchard C, Waldbaum A, et al. Low dose of transdermal estradiol (E2) gel for treatment of symptomatic postmenopausal women. Obstet Gynecol 2007;109(2):1–10.
  53. Montplaisir J, Lorrain J, Denesle R, et al. Sleep in menopause: differential effects of two forms of hormone replacement therapy. Menopause 2001;8(1):10–6.
  54. Gambacciani M, Ciaponi M, Cappagli B, et al. Effects of low-dose, continuous combined hormone replacement therapy on sleep in symptomatic postmenopausal women. Maturitas 2005;50:91–7.
  55. Zegura B, Guzic-Salobir B, Sebestjen M, et al. The effect of various menopausal hormone therapies on markers of inflammation, coagulation, fibrinolysis, lipids, and lipoproteins in healthy postmenopausal women. Menopause 2006;13(4): 643–50.
  56. Rossow J, Anderson G, Prentice R, et al. Writing Group for the Women’s Health Initiative. Risks and benefits of estrogen plus progestin in healthy postmenopausal women. JAMA 2002;288(3):321–33.
  57. Wassertheil-Smoller S, Hendrix S, Limacher M, et al. Effects of estrogen plus progestin on stroke in postmenopausal women. The women’s health initiative : a randomized trial. JAMA 2003;289(20):2673–84.
  58. Porch J, Lee I, Cook N, et al. Estrogen-progestin replacement therapy and breast cancer risk: the women’s health study (United States). Cancer Causes Control 2002;13:847–54.
  59. Statement on the estrogen plus progestin trial of the Women’s Health Initiative. ACOG News release 2002.
  60. HERS Study report. HT can relieve menopause-type symptoms common in elderly women. ACOG News Release 2002.
  61. Clarke C, Glaser S. Declines in breast cancer after the WHI: apparent impact of hormone therapy. Cancer Causes Control 2007;18(8):847–52.
  62. Fletcher S, Colditz GA. Failure of estrogen plus progestin therapy for prevention. JAMA 2002;288(3):366–8.
  63. Anderson G, Chlebowski R, Rossouw J, et al. Prior hormone therapy and breast cancer risk in the women’s health initiative randomized trial of estrogen plus progestin. Maturitas 2006;55:1–13.
  64. Lee S, Kolonel L, Wilkens L. Postmenopausal hormone therapy and breast cancer risk: the multiethnic cohort. Int J Cancer 2006;118:1285–91.
  65. Olsson H, Ingvar C, Bladstrom A. Hormone replacement therapy containing progestins and given continuously increases breast carcinoma risk in Sweden. Cancer 2003;97(6):1387–92.
  66. Jernstrom H, Bendahl P, Lidfeldt J, et al. A prospective study of different types of hormone replacement therapy use and the risk of subsequent breast cancer: the Women’s Health in the Lund Area (WHILA) study (Sweden). Cancer Causes Control 2003;14:673–80.
  67. Dolan S, Wilkie N. Arch effects of testosterone administration on human immunodeficiency virus-infected women with low weight. Arch Intern Med 2004;164: 897–904.
  68. Davis S, Walker K. Effects of estradiol with and without testosterone on body composition and relationship with lipids in postmenopausal women. Menopause 2000;7:395–401.
  69. Dobs A, Nguyen T. Differential effects of oral estrogen versus oral estrogen-androgen replacement therapy on body composition in postmenopausal women. Clin Endocrinol Metab 2002;87:1509–16.
  70. Miller K, Biller B, Beauregard C. Effects of testosterone replacement in androgen-deficient women with hypopituitarism; a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab 2006;91:1683–90.
  71. Davis S, Davidson S, Donath S. Circulating androgen levels and self-reported sexual function in women. JAMA 2005;294:91–6.
  72. Shifren J, Braunstein G, Simon J. Transdermal testosterone treatment in women with impaired sexual function after oophorectomy. NEJM 2000;343:682–8.
  73. Ando S, De Amicis F. Breast cancer from estrogen to androgen receptor. V Mol Molecular and Cellular Endocrinology 2002;193:121–8.
  74. Gammon M, Thompson W. Polycystic ovaries and the risk of breast cancer. Am J Epidemiol 1991;134:818–24.
  75. Winters J. Current status of testosterone replacement therapy in men. Arch Fam Med 1999;8:257–63.
  76. Mulligan T, Frick M, Zuraw Q, et al. Prevalence of hypogonadism in males aged at least 45 years: the HIM study. Int J Clin Pract 2006;60(7):762–9.
  77. Shippen E, Fryer W. The testosterone syndrome: the critical factor for energy, health and sexuality—reversing the male menopause. New York: M Evans and company; 1998.
  78. Khaw K, Dowsett M, Folkerd E, et al. Endogenous testosterone and mortality due to all causes, cardiovascular disease, and cancer in men. European Prospective Investigation into Cancer in Norfolk (EPIC-Norfolk) prospective population study. Circulation 2007;116:2694–701.
  79. Shores MM, Matsumoto AM, Sloan KL, et al. Low serum testosterone and mortality in male veterans. Arch Intern Med 2006;166:1660–5.
  80. English KM, Steeds RP, Jones HT, et al. Low-dose transdermal therapy improves angina theshold in men with chronic stable angina: a randomized, double-blind place-controlled study. Circulation 2000;102:1906–11.
  81. Kapoor D, Goodwin E, Channer KS, et al. Testosterone replacement therapy improves insulin resistance, glycaemic control, visceral adiposity and hypercholesterolaemia in hypogonadal men with type 2 diabetes. Eur J Endocrinol 2006; 154:899–906.
  82. Saad F, Gooren L, Haider A, et al. Effects of testosterone gel followed by parenteral testosterone undecanoate on sexual dysfunction and on features of the metabolic syndrome. Andrologia 2008;40:44–8.
  83. Allan CA, Strauss BJG, Burger HG, et al. Testosterone therapy prevents gain in visceral adipose tissue and loss of skeletal muscle in nonobese aging men. J Clin Endocrinol Metab 2008;93:139–46.
  84. Selvin E, Feinleib M, Zhang L, et al. Androgens and diabetes in men: results from the Third National Health and Nutrition Examination Survey (NHANES III). Diabetes Care 2001;30(2):234–8.
  85. Pitteloud N, Hardin M, Dwyer AA, et al. Increasing insulin resistance is associated with a decrease in Leydig cell testosterone secretion in men. J Clin Endocrinol Metab 2005;90:2636–41.
  86. Rodriguez A, Muller DC, Metter EJ, et al. Aging, androgens, and the metabolic syndrome in a longitudinal study of aging. J Clin Endocrinol Metab 2007;92(9): 3568–72.
  87. Ferrucci L, Maggio M, Bandinelli S, et al. Low testosterone predicts anemia in older adults. Arch Intern Med 2006;166:1380–8.
  88. Lu P, Masterman D. Effects of testosterone on cognition, and mood in male patients with mild Alzheimer disease and healthy elderly men. Arch Neurol 2006;63:177–85.
  89. Jackson J, Kleerekoper M. Osteoporosis in men; diagnosis, pathophysiology and prevention. Medicine 1990;69:137–52.
  90. Behre H, Kleisch S. Long term effect of testosterone therapy on bone mineral density in hypogonadal men. J Clin Endocrinol Metab 1997;82:2386–90.
  91. Herbst K, Bhasin S. Testosterone action on skeletal muscle. Curr Opin Clin Nutr Metab Care 2004;7(3):271–7.
  92. Davidson J, Camargo C. Effects of androgen on sexual behavior in hypogonadal men. J Clin Endocrinol Metab 1979;48:149–61.
  93. Shabsigh R, Kaufman J, Steidle C, et al. Randomized study of testosterone gel as adjunctive therapy to sildenafil in hypogonadal men with erectile dysfunction who do not respond to sildenafil alone. The Journal of Urology 2008;179(5): S97–102.
  94. Saad F, Grahl AS, Aversa A, et al. Effects of testosterone on erectile function: implications for the therapy of erectile dysfunction. BJU Int 2007;99(5): 988–92.
  95. Greco EA, Spera G, Aversa A. Combining testosterone and PDE5 inhibitors in erectile dysfunction: basic rationale and clinical evidences. Eur Urol 2006; 50(5):940–7.
  96. Huggins C, Hodges CV. Studies on prostatic cancer I: the effects of castration, of estrogen and of androgen injection on serum phosphotases in metastatic carcinoma of the prostate. Cancer Res 1941;1:293–7.
  97. Huggins C, Stevens RE, Hodges CV. Studies on prostatic cancer II: the effects of castration on advanced carcinoma of the prostate gland. Arch Surg 1941;43: 209–23.
  98. Morgentaler A. Testosterone replacement therapy and prostate cancer. Urol Clin North Am 2007;34:555–63.
  99. Harman S. Testosterone in older men after the Institute of Medicine report: Where do we go from here? Climacteric 2006;77(5):1319–26.
  100. Phillips G. Relationship between serum sex hormones and the glucose-insulin– lipid defect in men with obesity. Metabolism 1993;42(1):116–20.
  101. Carruthers M, Trinick TR, Wheeler MJ. The validity of androgen assays. The Aging Male 2007;10(3):165–72.
  102. Winters SJ. Endocrine evaluation of testicular function. Endocrinol Metab Clin North Am 1994;23:709–23.
  103. Lazarou S, Reyes-Vallejo L, Morgentaler A. Wide variability in laboratory reference values for serum testosterone. J Sex Med 2006;3:1085–9.
  104. Travison T, Araujo AB, O’Donnell AB, et al. A population-level decline in serum testosterone levels in American men. J Clin Endocrinol Met 2007;92(1): 196–202.
  105. Skakkebæk NE, Jørgensen N, Main KM, et al. Is human fecundity declining? J Androl 2006;29(1):2–11.
  106. Matsumoto AM. Hormonal therapy of male hypogonadism. Endocrinol Metab Clin North Am 1994;23:857–75.
  107. Carter HB, Ferrucci L, Kettermann A, et al. Detection of life-threatening prostate cancer with prostate-specific antigen velocity during a window of curability. J Natl Cancer Inst 2006;98(21):1521–7.
  108. Morgentaler A. Commentary: guidelines for male testosterone therapy: a clinician’s perspective. J Clin Endocrinol Metab 2007;92(2):416–7.
  109. Maghnie M, Aimaretti G, Bellone S, et al. Diagnosis of GH deficiency in the transition period: accuracy of insulin tolerance test and insulin-like growth factor-I measurement. European Journal of Endocrinology 2005;152(4):589–96.
  110. Gudman D, Kutner MH, Rogers M, et al. Impaired growth hormone secretion in the adult population. J clin Invest 1981;67:1361–9.
  111. Zadik Z, Chalew SA, McCarter RJ, et al. The influence of age on the 24-hour integrated concentration of growth hormone in normal individuals. J Clin Endocrinol Metab 1985;60:513–6.
  112. Ho KY, Evans WS, Blizzard RM, et al. Effects of sex and age on the 24-hour profile of growth hormone secretion in man: importance of endogenous estradiol concentrations. J Clin Endocrinol Metab 1987;64:51–8.
  113. McGauley G. The psycologogical consequences and quality of life in adults with hormone deficiency. Growth Horm IGF Res 2000;10:S63–8.
  114. Tiryakioaylu O, Kadiolgu P, Canerolgu NU, et al. Age dependency of serum insulin-like growth factor (IGF)-1 in healthy Turkish adolescents and adults. Indian J Med Sci 2003;57(12):543–8.
  115. Savine R, Sonksen P. Growth hormone-hormone replacement for the somatopause. Horm Res 2000;53(3):37–41.
  116. Deijen JB, van der Veen EA. The influence of growth hormone (GH) deficiency and GH replacement on quality of life in GH-deficient patients. J Endocrinol Invest 1999;22(5 Suppl):127–36.
  117. Murray RD, Darzy KH, Gleeson HK, et al. GH-deficient survivors of childhood cancer: GH replacement during adult life. J Clin Endocrinol Metab 2002;87(1):129–35.
  118. Wureb L, Bengtsson BA, Johannsson G. Beneficial effects of long-term GH replacement therapy on quality of life in adults with GH deficiency. Clin Endocrinol 1998;48:613–20.
  119. Gibney J, Wallace JD, Spinks T, et al. The effects of 10 years of recombinant human growth hormone (GH) in adult GH-deficient patients. J Clin Endocrinol Metab 1999;84(8):2596–602.
  120. Bennett RM, Clark SC, Walczyk J. A randomized, double-blind, placebocontrolled study of growth hormone in the treatment of fibromyalgia. Am J Med 1998;104(3):227–31.
  121. Johannsson G, Svensson J, Bengtsson BA. Growth hormone and ageing. Growth Horm IGF Res 2000;10(2):25–30.
  122. Maison P, Philippe C. Cardiac effects of growth hormone in adults with growth hormone deficiency: a meta-analysis. Circulation 2003;108:2648–52.
  123. Cho GY, Jeong IK, Kim SH, et al. Effect of growth hormone on cardiac contractility in patients with adult onset growth hormone deficiency. Amerasia J 2007; 100(6):1035–9.
  124. Johannsson G, Marin P, Lonn L, et al. GH treatment of abdominally obese men reduces abdominal fat mass, improves glucose and lipoprotein metabolism and reduces diastolic BP. J Clin Endocrinol Metab 1997;82:727–34.
  125. Gotherstrom G, et al. A prospective study of 5 years of GH replacement therapy in GH-deficient adults: sustained effects on body composition, bone mass, and metabolic indices. J Clin Endocrinol Metab 2001;86(10):4657–65.
  126. Feldt-Rasmussen B, Lange M, Sulowicz W, et al. Growth hormone treatment during hemodialysis in a randomized trial improves nutrition, quality of life, and cardiovascular risk. J Am Soc Nephrol 2007;18(7):2161–71.
  127. Yuen KC, Dunger DB. Impact of treatment with recombinant human GH and IGF-1 on visceral adipose tissue and glucose homeostasis in adults. Growth Horm IGF Res 2006;16:S55–61.
  128. Aleman A, Verhaar HJ, de Haan EH, et al. Insulin-like growth factor-1 and cognitive function in healthy older men. J Clin Endocrinol Metab 1999;84: 471–5.
  129. Arwert LI, Veltman DJ, Deijen JB, et al. Effects of growth hormone substitution therapy on cognitive functioning in growth hormone deficient patients: a functional MRI study. Neuroendocrinology 2006;83:12–9.
  130. Clark R. The somatogenic hormones and insulin-like growth factor-1: stimulators of lymphopoiesis and immune function. Endocr Rev 1997;18(2):157–79.
  131. Burgess W, Liu Q, Jian-Hua Zhou J, et al. The immune-endocrine loop during aging: role of growth hormone and insulin-like growth factor-1. Neuroimmunomodulation 1999;6(1-2):56–68.
  132. Rudman D. Effects of growth hormone in men over 60 years old. N Engl J Med 1990;323(1):1–6.
  133. Munzer T, Harman SM, Hees P, et al. Effects of GH and/or sex steroid administration on abdominal subcutaneous and visceral fat in healthy aged women and men. J Clin Endocrinol Metab 2001;86(8):3604–10.
  134. Pfeifer M, Verhovec R, Zizek B, et al. Growth hormone (GH) treatment reverses early atherosclerotic changes in GH-deficient adults. J Clin Endocrinol Metab 1999;84:453–7.
  135. Borson-Chazot F, Serusclat A, Kalfallah Y, et al. Decrease in carotid intimamedia thickness after one year growth hormone (GH) treatment in adults with GH deficiency. J Clin Endocrinol Metab 1999;84:1329–33.
  136. Valimaki MJ, Salmela PI, Salmi J, et al. Effects of 42 months of GH treatment on bone mineral density and bone turnover in GH-deficient adults. Eur J Endocrinol 1999;140(6):545–54.
  137. Maghnie M, Aimaretti G, Bellone S, et al. Diagnosis of GH deficiency in the transition period: accuracy of insulin tolerance test and insulin-like growth factor-1 measurement. Eur J Endocrinol 2005;152(4):589–96.
  138. Hoeck HC, Vestergaard P, Jakobsen PE, et al. Test of growth hormone secretion in adults: poor reproducibility of the insulin tolerance test. Eur J Endo 1995;133: 305–12.
  139. Hoeck HC, Jakobsen PR, Vestergaard P, et al. Differences in reproducibility and peak growth hormone responses to repeated testing with various stimulators in healthy adults. Growth Horm IGF Res 1999;9:18–24.
  140. Rahim A, Toogood AA, Shalet SM. The assessment of growth hormone status in normal young adult males using a variety of provocative agents. Clin Endo 1996; 45:557–62.
  141. Cacciari E, Cicognani A, Pirazzoli P, et al. Differences in somatomedin-C between short-normal subjects and those of nomal height. J Pediatr 1985; 106:891–4.
  142. Wilson DM, Frane J. A brief review of the use and utility of growth hormone stimulation testing in the NCGS: do we need to do provocative GH testing? Growth Hormone & IGF Research 2005;15:S21–5.
  143. Tassoni P, Cacciari E, Cau M, et al. Variability of growth hormone response to pharmacological and sleep tests performed twice in short children. J Clin Endocrinol Metab 1990;71(1):230–4.
  144. Gandrud LM, Wilson DM. Is growth hormone stimulation testing in children still appropriate? Growth Hormone & IGF-1 Research 2004;14:185–94.
  145. Shah A, Stanhope R, Matthew D. Hazards of pharmacological tests of growth hormone secretion in childhood. BMJ 1992;304:173–4.
  146. Wuster C, Melchinger U, Eversmann T, et al. Reduced incidence of side-effects of growth hormone substitution in 404 patients with hypophyseal insufficiency. Results of a multicenter indications study. Med Klin 1998;93(10):585–91.
  147. Amato G, Izzo G, La Montagna G, et al. Low dose recombinant human growth hormone normalizes bone metabolism and cortical bone density and improves trabecular bone density in growth hormone deficient adults without causing adverse effects. Clin Endocrinol (Oxf) 1996;45(1):27–32.
  148. Chihara K, Koledova E, Shimatsu A, et al. An individualized GH dose regimen for long-term GH treatment in Japanese patients with adult GH deficiency. Eur J Endocrinol 2005;153(1):57–65.
  149. Hankinson SE, Willett WC, Colditz GA, et al. Circulating concentrations of insulin-like growth factor-1 and risk of breast cancer. Lancet 1998;351(9113): 1393–8.
  150. Chan JM, Stampfer MJ, Giovannucci E, et al. Plasma insulin-like growth factor-1 and prostate cancer risk: a prospective study. Science 1998;279(23):563–6.
  151. Palmqvist R, Hallmans G, Rinaldi S, et al. Plasma insulin-like growth factor 1, insulin-like growth factor binding protein 3, and risk of colorectoal cancer: a prospective study in northern Sweden. Gut 2002;50:642–6.
  152. Agurs-Collins T, Adams-Campbell LL, Kim KS, et al. Insulin-like growth factor-1 and breast cancer risk in postmenopausal African-American women. Cancer Detect Prev 2000;24(3):199–206.
  153. Baffa R, Reiss K, El-Gabry EA, et al. Low serum insulin-like growth factor 1 (IGF-1): a significant association with prostate cancer. Tech Urol 2000;6(3):236–9.
  154. Spitz MR, Barnett MJ, Goodman GE, et al. Serum insulin-like growth factor (IGF) and IGF-binding protein levels and risk of lung cancer: a case-control study nested in the beta-carotene and retinol efficacy trial cohort. Cancer Epidemiol Biomarkers Prev 2002;11(11):1413–8.
  155. Kurek R, Tunn UW, Eckart O, et al. The significance of serum levels of insulin-like growth factor-1 in patients with prostate cancer. BJU Int 2000;85(1):125–9.
  156. Cutting CW, Hunt C, Nisbet JA, et al. Serum insulin-like growth factor-1 is not a useful marker of prostate cancer. BJU Int 1999;83(9):996–9.
  157. Fuhrman B, Barba M, Schu¨nemann HJ, et al. Basal growth hormone concentrations in blood and the risk for prostate cancer: a case control study. Prostate 2005;64(2):109–15.
  158. Chen C, Lewis SK, Voigt L, et al. Prostate carcinoma incidence in relation to prediagnostic circulating levels of insulin-like growth factor I, insulin like growth factor binding protein 3, and insulin. Cancer 2005;103(1):76–84.
  159. Li BD, Khosravi MJ, Berkel HJ, et al. Free insulin-like growth factor-I and breast cancer risk. Int J Cancer 2001;91(5):736–9.
  160. Lo¨n S, Inskip PD, Pollak MN, et al. Glioma risk in relation to serum levels of insulinlike growth factors. Cancer Epidemiol Biomarkers Prev 2007;16(4):844–6.
  161. Finne P, Auvinen A, Koistinen H, et al. Insulin-like growth factor I is not a useful marker of prostate cancer in men with elevated levels of prostate-specific antigen. J Clin Endocrinol Metab 2000;85(8):2744–7.
  162. Woodson K, Tangrea JA, Pollak M, et al. Serum IGF-1: tumor marker or etiologic factor? A prospective study of prostate cancer among Finnish men. Cancer Res 2003;63(14):3991–4.
  163. Lacey JV Jr, Potischman N, Madigan MP, et al. Insulin-like growth factors, insulin-like growth factor-binding proteins, and endometrial cancer in postmenopausal women: results from a U.S. case-control study. Cancer Epidemiol Biomarkers Prev 2004;13(4):607–12.
  164. Schaffer A, Koushik A, Trottier H, et al. Biomarkers of cervical cancer risk study team. Insulin-like growth factor-I and risk of high-grade cervical intraepithelial neoplasia. Cancer Epidemiol Biomarkers Prev 2007;16(4):716–22.
  165. Serrano ML, Romero A, Cendales R, et al. Serum levels of insulin-like growth factor-I and -II and insulin-like growth factor binding protein 3 in women with squamous intraepithelial lesions and cervical cancer. Biomedica 2006;26(2): 258–68.
  166. Karavitaki N, Warner JT, Marland A, et al. GH replacement does not increase the risk of recurrence in patients with craniopharyngioma. Clin Endocrinol (Oxf) 2006;64(5):556–60.
  167. Buchfelder M, Kann PK, Wuster C, et al. Influence of GH substitution therapy in deficient adults on the recurrence rate of hormonally inactive pituitary adenomas: a case control study. European journal of endocrinology 2007;157(2):149–56.
  168. Smit P, Koppeschaar H. Growth hormone therapy and risk of malignancy. Endocrinologist 2008;18(1):39–43.
  169. Swerdlow AJ, Reddingius RE, Higgins CD, et al. Growth hormone treatment of children with brain tumors and risk of tumor recurrence. J Clin Endocrinol Metab 2000;85(12):4444–9.
  170. Tacke J, Bolder U, Herrmann A, et al. Long-term risk of gastrointestinal tumor recurrence after postoperative treatment with recombinant human growth hormone. J Parenter Enteral Nutr 2000;24(3):140–4.
  171. Critical evaluation of the safety of recombinant human growth hormone administration: statement from the Growth Hormone Research Society. J Clin Endocrinol Metab 2001;86:1868–70.
  172. Hormones in Wellness and Disease Prevention 699 172. Svensson J, Bengtsson BA˚ , Rosen T, et al. Malignant disease and cardiovascu- lar morbidity in hypopituitary adults with or without hormone replacement therapy. J Clin Endocrinol Metab 2004;89(7):3306–12.
  173. Shalet SM, Brennan BM, Reddingius RE. Growth hormone therapy and malignancy. Horm Res 1997;48(Suppl 4):29–32.
  174. Pollak M. Insulin-like growth factors and prostate cancer. Epidemiol Rev 2001; 23(1):59–66.
  175. Leung W, Zhou Y, Hancock ML, et al. Outcomes of growth hormone replacement therapy in survivors of childhood acute lymphoblastic leukemia. J Clin Oncol 2002;20(13):2959–64.
  176. Blethen SL, Allen DB, Graves D, et al. Safety of recombinant deoxyribonucleic acid–derived growth hormone: the national cooperative growth study experience. J Clin Endocrinol Metab 1996;81:1704–10.
  177. Frajese G, Drake WM, Loureiro RA, et al. Hypothalamopituitary surveillance imaging in hypopituitary patients receiving long-term GH replacement therapy. J Clin Endocrinol Metab 2001;86(11):5572–5.
  178. Blackman M, Sorkin DJ, Munzer T, et al. Growth hormone and sex steroid administration in healthy aged women and men: a randomized controlled trial. JAMA 2002;288(18):2282–92.
  179. Yuen KC, Frystyk J, White DK, et al. Improvement in insulin sensitivity without concomitant changes in body composition and cardiovascular risk markers following fixed administration of a very low growth hormone (GH) dose in adults with severe GH deficiency. Clin Endocrinol (Oxf) 2005;63(4):428–36.
  180. Yuen KC, Dunger DB. Impact of treatment with recombinant human GH and IGF-1 on visceral adipose tissue and glucose homeostasis in adults. Growth Horm IGF Res 2006;16:S55–61.
  181. Svensson J, Fowelin J, Landin K, et al. Effects of seven years of GHreplacement therapy on insulin sensitivity in GH-deficient adults. J Clin Endocrinol Metab 2002;87:2121–7.
  182. Ahn C, Kim C, Nam J, et al. Effects of growth hormone on insulin resistance and atherosclerotic risk factors in obese type 2 diabetic patients with poor glycaemic control. Clin Endocrinol 2006;64:444–9.
  183. Canaris GJ, Manowitz NR, Mayor G, et al. The Colorado thyroid disease prevalence study. Arch Intern Med 2000;160:526–34.
  184. Mcdermott MT, Ridgway C. Subclinical hypothyroidism is mild thyroid failure and should be treated. J Clin Endocrinol Met 2001;86(10):4585–90.
  185. Monzani F, Del GuerraP, CaraccioN,et al. Subclinical hypothyroidism: neurobehavioral features and beneficial effect of l-thyroxine treatment. Clin Invest 1993;71:367–71.
  186. Tappy L, Randin JP, Schwed P, et al. Prevalence of thyroid disorders in psychogeriatric inpatients. A possible relationship of hypothyroidism with neurotic depression but not dementia. J Am Geriatr Soc 1987;35:526–31.
  187. Joffe RT, Levitt AJ. Major depression and subclinical (grade 2) hypothyroidism. Psychoneuroendocrinology 1992;17:215–21.
  188. Haggerty JJ Jr, Stern RA, Mason GA, et al. Subclinical hypothyroidism: a modifiable risk factor for depression? Am J Psychiatry 1993;150(3):508–10.
  189. Manciet G, Dartigues JF, Decamps A, et al. The PAQUID survey and correlates of subclinical hypothyroidism in elderly community residents in the southwest of France. Age Ageing 1995;24:235–41.
  190. Baldini IM, Vita A, Maura MC, et al. 1997 Psychological and cognitive features in subclinical hypothyroidism. Prog Neurophychopharmacol Biol Psychiatry 1997; 21:925–35.
  191. Ganguli M, Burmeister LA, Seaberg EC, et al. Association between dementia and elevated TSH: a community-based study. Biol Psychiatry 1996;40:714–25.
  192. Monzani F, Caraccio N, Siciliano G, et al. Clinical and biochemical features of muscle dysfunction in subclinical hypothyroidism. J Clin Endocrinol Metab 1997;82:3315–8.
  193. Monzani F, Caraccio N, Del Guerra P, et al. Neuromuscular symptoms and dysfunction in subclinical hypothyroid patients: beneficial effect of L-T4 replacement therapy. Clin Endocrinol 1999;51:237–42.
  194. Misiunas A, Ravera HN, Faraj G, et al. Peripheral neuropathy in subclinical hypothyroidism. Thyroid 1995;5:283–6.
  195. Goulis DG, Tsimpiris N, Delaroudis S, et al. Stapedial reflex: a biological index found to be abnormal in clinical and subclinical hypothyroidism. Thyroid 1998; 8:583–7.
  196. Ridgway EC, Cooper DS, Walker H, et al. Peripheral responses to thyroid hormone before and after L-thyroxine therapy in patients with subclinical hypothyroidism. J Clin Endocrinol Metab 1981;53:1238–42.
  197. Cooper DS, Halpern R, Wood LC, et al. l-thyroxine therapy in subclinical hypothyroidism. Ann Intern Med 1984;101:18–24.
  198. Nystrom E, Caidahl K, Fager G, et al. A double-blind cross-over 12-month study of L-thyroxine treatment of women with ‘subclinical’ hypothyroidism. Clin Endocrinol 1988;29:63–76.
  199. Bell GM, Todd WT, Forfar JC, et al. End-organ responses to thyroxine therapy in subclinical hypothyroidism. Clin Endocrinol (Oxf) 1995;22:83–9.
  200. Forfar JC, Wathen CG, Todd WT, et al. Left ventricular performance in subclinical hypothyroidism. QJM 1985;57:857–65.
  201. Foldes J, Istvanfy M, Halmagyi M, et al. Hypothyroidism and the heart. Examination of left ventricular function in subclinical hypothyroidism. Acta Med Hung 1987;44:337–47.
  202. Kahaly GJ. Cardiovascular and atherogenic aspects of subclinical hypothyroidism. Thyroid 2000;10:665–79.
  203. Monzani F, Di Bello V, Caraccio N, et al. Effect of levothyroxine on cardiac function and structure in subclinical hypothyroidism: a double blind, placebocontrolled study. J Clin Endocrinol Metab 2001;86:1110–5.
  204. Hak EA, Pols HA, Visser TJ, et al. Subclinical hypothyroidism is an independent risk factor for atherosclerosis and myocardial infarction in elderly women: the Rotterdam study. Ann Intern Med 2000;4:270–8.
  205. Tanis BC, Westendorp RGJ, Smelt AHM. Effect of thyroid substitution on hypercholesterolaemia in patients with subclinical hypothyroidism: a re-analysis of intervention studies. Clin Endocrinol 1996;44:643–9.
  206. Danese MD, Ladenson PW, Meinert CL, et al. Effect of thyroxine therapy on serum lipoproteins in patients with mild thyroid failure: a quantitative review of the literature. J Clin Endocrinol Metab 2000;85:2993–3001.
  207. Michalopoulou G, Alevizaki M, Piperingos G, et al. High serum cholesterol levels in persons with ‘high normal’ TSH levels: Should one extend the definition of subclinical hypothyroidism. Eur J Endocrinol 1998;138:141–5.
  208. Bindels AJ, Westendorp RG, Frolich M, et al. The prevalence of subclinical hypothyroidism at different total plasma cholesterol levels in middle aged men and women: a need for case-finding? Clin Endocrinol 1999;50:217–20.
  209. Bakker SJL, Ter Matten JC, Popp-Snijders C, et al. The relationship between thyrotropin and low density lipoprotein cholesterol is modified by insulin sensitivity in healthy euthyroid subjects. J Clin Endocrinol Metab 2001;86:1206–11.
  210. Lekakis J, Papamichael C, Alevizaki M, et al. Flow-mediated, endotheliumdependent vasodilatation is impaired in subjects with hypothyroidism, borderline hypothyroidism, and high-normal serum thyrotropin (TSH) values. Thyroid 1997;7:411–4.
  211. Walsh JP, Bremner AP, Bulsara MK, et al. Subclinical thyroid dysfunction as a risk factor for cardiovascular disease. Arch Intern Med 2005;165(21):2467–72.
  212. Peeters RP, Geyten SV, Wouters PJ, et al. Tissue thyroid hormone levels in critical illness. J Clin Endocrinol Metab 2005;12:6498–507.
  213. Docter R, Krenning EP, de Jong M, et al. The sick euthyroid syndrome: changes in thyroid hormone serum parameters and hormone metabolism. Clin Endocrinol (Oxf) 1993;39:499–518.
  214. Fliers E, Alkemade A, Wiersinga WM. The hypothalamic-pituitary-thyroid axis in critical illness. Best Practice & Research Clinical Endocrinology & Metabolism 2001;15(4):453–64.
  215. Chopra IJ. Euthyroid sick syndrome: Is it a misnomer? J Clin Endocrinol Metab 1997;82(2):329–34.
  216. van der Poll T, Romijn JA, Wiersinga WM, et al. Tumor necrosis factor: a putative mediator of the sick euthyroid syndrome in man. J Clin Endocrinol Metab 1990; 71(6):1567–72.
  217. Stouthard JM, van der Poll T, Endert E, et al. Effects of acute and chronic interleukin-6 administration on thyroid hormone metabolism in humans. J Clin Endocrinol Metab 1994;79(5):1342–6.
  218. Corssmit EP, Heyligenberg R, Endert E, et al. Acute effects of interferon-alpha administration on thyroid hormone metabolism in healthy men. Clin Endocrinol Metab 1995;80(11):3140–4.
  219. Nagaya T, Fujieda M, Otsuka G, et al. A potential role of activated NF-Kappa B in the pathogenesis of euthyroid sick syndrome. J Clin Invest 2000;106(3): 393–402.
  220. Bianco AC, Salvatore D, Gereben B, et al. Biochemistry, cellular and molecular biology, and physiological roles of the iodothyronine selenodieidinases. Endocr Rev 2002;23:38–89.
  221. Chopra IJ, Huang TS, Beredo A, et al. Evidence for an inhibitor of extrathyroidal conversion of thyroxine to 3,5,3’-triiodothyronine in sera of patients with nonthyroidal illnesses. J Clin Endocrinol Metab 1985;60:666–72.
  222. Peeters RP, Wouters PJ, Kaptein E, et al. Reduced activation and increased inactivation of thyroid hormone in tissues of critically ill patients. J Clin Endocrinol Metab 2003;88:3202–11.
  223. Chopra IJ, Chopra U, Smith SR, et al. Reciprocal changes in serum concentrations of 3,3’,5-triiodothyronine (T3) in systemic illnesses. J Clin Endocrinol Metab 1975;41:1043–9.
  224. Iervasi G, Pinitore A, Landi P, et al. Low-T3 syndrome a strong prognostic predictor of death in patients with heart disease. Circulation 2003;107(5): 708–13.
  225. Peeters RP, Wouters PJ, van Toor H, et al. Serum 3,3’,5’-triiodothyronine (rT3) and 3,5,3’-triiodothyronine/rT3 are prognostic markers in critically ill patients and are associated with postmortem tissue deiodinase activities. J Clin Endocrinol Metab 2005;90(8):4559–65.
  226. Wartofsky L, Burman K. Alterations in thyroid function in patients with systemic illness; the ‘‘euthyroid sick syndrome’’. Endocr Rev 1982;3(2):164–217.
  227. Hennemann G, Everts ME, de Jong, et al. The significance of plasma membrane transport in the bioavailability of thyroid hormone. Clin Endocrinol 1998;48:1–8.
  228. Vos RA, de Jong M, Bernard HF, et al. Impaired thyroxine and 3,5,3’-triodothyronine handling by rat hepatocytes in the presence of serum of patients with nonthryoidal illness. J Clin Endocrinology met 1995;80:2364–70.
  229. Chopra IJ, Solomon DH, Hepner GW, et al. Misleadingly low free thyroxine index and usefulness of reverse triiodothyronine measurement in nonthyroidal illnesses. Ann Intern Med 1979;90(6):905–12.
  230. De Jong M, Docter R, Van Der Hoek HJ, et al. Transport of 3,5,3’-triiodothyronine into the perfused rat liver and subsequent metabolism are inhibited by fasting. Endocrinology 1992;131:463–70.
  231. Mooradian AD, Reed RL, Osterweil D, et al. Decreased serum triiodothyronine is associated with increased concentrations of tumor necrosis factor. J Clin Endocrinol Metab 1990;71(5):1239–42.
  232. de Jong F, den Heijer T, Visser TJ, et al. Thyroid hormones, dementia, and atrophy of the medical temporal lobe. J Clin Endocrinol Met 2006;91(7): 2569–73.
  233. Carrero JJ, Qureshi AR, Axelsson J, et al. Clinical and biochemical implications of low thyroid hormone levels (total and free forms) in euthyroid patients with chronic kidney disease. J Intern Med 2007;262:690–701.
  234. Zoccali C, Tripepi G, Cutrupi S, et al. Low triiodothyronine: a new facet of inflammation in end-stage renal disease. J Am Soc Nephrol 2005;16:2789–95.
  235. Zoccali C, Mallamaci F, Tripepi G, et al. Low triiodothyronine and survival in endstage renal disease. Kidney Int 2006;70:523–8.
  236. Naslund E, Andersson I, Degerblad M, et al. Associations of leptin, insulin resistance and thyroid function with long-term weight loss in dieting obese men. J Int Med 2000;248:299–308.
  237. Pingitore A, Landi P, Taddei MC, et al. Triiodothyronine levels for risk stratification of patients with chronic heart failure. Am J Med 2005;118(2):132–6.
  238. Kozdag G, Ural D, Vural A, et al. Relation between free triiodothyronine/free thyroxine ratio, echocardiographic parameters and mortality in dilated cardiomyopathy. Eur J Heart Fail 2005;7(1):113–8.
  239. Karadag F, Ozcan H, Karul AB, et al. Correlates of non-thyroidal illness syndrome in chronic obstructive pulmonary disease. Respir Med 2007;101: 1439–46.
  240. Kok P, Roelfsema F, Langendonk JG, et al. High circulating thyrotropin levels in obese women are reduced after body weight loss induced by caloric restriction. J Clin Endocrinol Metab 2005;90:4659–63.
  241. Ohyama T, Aono T, Nakai A, et al. Circulation free T3 in pregnancy, liver disease, diabetes mellitus and thyroid disease. Nippon Naibunpi Gakkai Zasshi 1984;60: 1227–34.
  242. Parr JH. The effect of long-term metabolic control on free thyroid hormone levels in diabetics during insulin treatment. Ann Clin Biochem 1987;24(5):466–9.
  243. Dimopoulou I, Ilias I, Mastorakos G, et al. Effects of severity of chronic obstructive pulmonary disease on thyroid function. Metabolism 2001;50(12):1397–401.
  244. Mariotti S, Barbesino G, Caturegli P, et al. Complex alterations of thyroid function in healthy centenarians. J Clin Endocrinol Met 1993;77(5):1130–4.
  245. Nomura S, Pittman CS, Chambers JB, et al. Reduced peripheral conversion of thyroxine to triiodothyronine in patients with hepatic cirrhosis. J Clin Invest 1975; 56:643–8.
  246. Pingitore A, Galli E, Barison A, et al. Acute effects of triiodothyronine replacement therapy in patients with chronic heart failure and low T3 syndrome: a randomized placebo-controlled study. J Clin Endocrinol Met 2008;93:1351–8.
  247. Dulchavsky SA, Kennedy PR, Geller ER, et al. T3 preserves respiratory function in sepsis. J Trauma 1991;31:753–9.
  248. Hesch RD, Husch M, Kodding R, et al. Treatment of dopamine-dependent shock with triiodothyronine. Endocr Res Commun 1981;8:299–301.
  249. Dulchavsky SA, Hendrick SR, Dutta S. Pulmonary biophysical effects of triidothyronine (T3) augmentation during sepsis-induced hypothyroidism. J Trauma 1993;35:104–9.
  250. Meyer T, Husch M, van den Berg E, et al. Treatment of dopamine-dependent shock with triiodothyronine: preliminary results. Dtsch Med Wochenschr 1979; 104:1711–4.
  251. Novitzky D, Cooper DK, Reichart B. Hemodynamic and metabolic responses to hormonal therapy in brain-dead potential organ donors. Transplantation 1987; 43:852–5.
  252. Dulchavsky SA, Maitra SR, Maurer J, et al. Beneficial effects of thyroid hormone administration on metabolic and hemodynamic function in hemorrhagic shock. FASEB J 1990;4:A952.
  253. Hamilton MA, Stevenson LW, Fonarow GC, et al. Safety and hemodynamic effects of intravenous triiodothyronine in advanced congestive heart failure. Am J Cardiol 1998;81:443–7.
  254. Klemperer JD, Klein IL, Ojamaa K, et al. Triidothyronine therapy lowers the incidence of atrial fibrillation after cardiac operations. Ann Thorac Surg 1996;61: 1323–9.
  255. Smidt-Ott UM, Ascheim DD. Thyroid hormone and heart failure. Curr Heart Fail Rep 2006;3:114–9.
  256. Carle A, Laurberg P, Pedersen IB, et al. Thyrotropin secretion decreases with age in patients with hypothyroidism. Clinical Thyroidology 2007;17:139–44.
  257. van den Beld AW, Visser TJ, Feelders RA, et al. Thyroid hormone concentrations, disease, physical function and mortality in elderly men. J Clin Endocrinol Metab 2005;90(12):6403–9.
  258. Hermann J, Heinen E, Kroll HJ, et al. Thyroid function and thyroid hormone metabolism in elderly people low T3–syndrome in old age. Klin Wochenschr 1981; 59:315–23.
  259. Fukagawa NK, Bandini LG, Young JB. Effect of age on body composition and resting metabolic rate. Am J Physiol Endocrinol Metab 1990;259:E233–8.
  260. van Coevorden A, Laurent E, Decoster C, et al. Decreased basal and stimulated thyrotropin secretion in healthy elderly men. J Clin Endocrinol Metab 1989;69: 177–85.
  261. Rubenstein HA, Butler VPJ, Werner SC. Progressive decrease in serum triiodothyronine concentrations with human aging: radioimmunoassay following extraction of serum. J Clin Endocrinol Metab 1973;37:247–53.
  262. Chakraborti S, Chakraborti T, Mandal M, et al. Hypothalamic–pituitary–thyroid axis status of humans during development of ageing process. Clin Chim Acta 1999;288(1-2):137–45.
  263. Piers LS, Soars MJ, McCormack LM, et al. Is there evidence for an age-related reduction in metabolic rate? J Appl Phys 1998;85:2196–204.
  264. Poehlman ET, Berke EM, Joseph JR, et al. Influence of aerobic capacity, body composition, and thyroid hormones on the age-related decline in resting metabolic rate. Metabolism 1992;41:915–21.
  265. Magri F, Fioravanti CM, vignati G, et al. Thyroid function in old and very old healthy subjects. J Endocrinol Invest 2002;25(10):60–3.
  266. Schwartz & Holtorf 266. Goichot B, Schlienger JL, Grunenberger F, et al. Thyroid hormone status and nutrient intake in the free-living elderly. Interest of reverse triiodothyronine assessment. Eur J Endocrinol 1994;130:244–52.
  267. Cizza G, Brady LS, Calogero AE, et al. Central hypothyroidism is associated with advanced age in male Fischer 344/n rats: in vivo and in vitro studies. Endocrinology 1992;131:2672–80.

More Articles

Article

Nanotechnology’s Latest Oncolytic Agent: Silver, Cancer & Infection Associations Part III

It has long been suspected that infectious agents are associated with solid tumor cancers (a notable example is Kaposi’s Sarcoma) as well as non-tumor based cancers such as leukemia.

Read More...
Article

Peripheral Thyroid Hormone Conversion - Impact on TSH and Metabolic Activity

There have been recent advances in understanding of the local control of thyroid activity and metabolism.

Read More...
Article

Hormone Replacement Therapy in the Geriatric Patient Part 1

Therapies for aging patients focus primarily on prolonging life, often at very high emotional and financial cost with little focus on the quality of life the patient experiences.

Read More...
Article

Hormones in Wellness and Disease Prevention

The study and use of hormones have long been the domains of endocrinology, which is primarily focused on the pathologic phenomena encountered in the human body as they relate to hormones.

Read More...

Quicklinks

AboutMedical LiteraturePublications

NEWSLETTER

Submit your email address below to stay up to date on the latest in healthcare, delivered straight to your inbox.

By submitting this form, you consent to receive marketing and promotional emails from Holtorf Medical Group. You may unsubscribe from this list at any time.
Terms of ServicePrivacy Policy